1
|
Meza-Torres J, Tinevez JY, Crouzols A, Mary H, Kim M, Hunault L, Chamorro-Rodriguez S, Lejal E, Altamirano-Silva P, Groussard D, Gobaa S, Peltier J, Chassaing B, Dupuy B. Clostridioides difficile binary toxin CDT induces biofilm-like persisting microcolonies. Gut Microbes 2025; 17:2444411. [PMID: 39719371 DOI: 10.1080/19490976.2024.2444411] [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: 07/19/2024] [Revised: 11/07/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
Clinical symptoms of Clostridioides difficile infection (CDI) range from diarrhea to pseudomembranous colitis. A major challenge in managing CDI is the high rate of relapse. Several studies correlate the production of CDT binary toxin by clinical strains of C. difficile with higher relapse rates. Although the mechanism of action of CDT on host cells is known, its exact contribution to CDI is still unclear. To understand the physiological role of CDT during CDI, we established two hypoxic relevant intestinal models, Transwell and Microfluidic Intestine-on-Chip systems. Both were challenged with the epidemic strain UK1 CDT+ and its isogenic CDT- mutant. We report that CDT induces mucin-associated microcolonies that increase C. difficile colonization and display biofilm-like properties by enhancing C. difficile resistance to vancomycin. Importantly, biofilm-like microcolonies were also observed in the cecum and colon of infected mice. Hence, our study shows that CDT induces biofilm-like microcolonies, increasing C. difficile persistence and risk of relapse.
Collapse
Affiliation(s)
- Jazmin Meza-Torres
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Jean-Yves Tinevez
- Image Analysis Hub, Department of Cell Biology and Infection, Institut Pasteur, Université Paris Cité, Paris, France
| | - Aline Crouzols
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Minhee Kim
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Lise Hunault
- Antibodies in Therapy and Pathology, Department of Immunology, Institut Pasteur, Paris, France
| | - Susan Chamorro-Rodriguez
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Emilie Lejal
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Pamela Altamirano-Silva
- Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | | | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Johann Peltier
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Department of Microbiology, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Bruno Dupuy
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| |
Collapse
|
2
|
Clyne M, Ó Cróinín T. Pathogenicity and virulence of Helicobacter pylori: A paradigm of chronic infection. Virulence 2025; 16:2438735. [PMID: 39725863 DOI: 10.1080/21505594.2024.2438735] [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: 05/13/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Infection with Helicobacter pylori is one of the most common infections of mankind. Infection typically occurs in childhood and persists for the lifetime of the host unless eradicated with antimicrobials. The organism colonizes the stomach and causes gastritis. Most infected individuals are asymptomatic, but infection also causes gastric and duodenal ulceration, and gastric cancer. H. pylori possesses an arsenal of virulence factors, including a potent urease enzyme for protection from acid, flagella that mediate motility, an abundance of outer membrane proteins that can mediate attachment, several immunomodulatory proteins, and an ability to adapt to specific conditions in individual human stomachs. The presence of a type 4 secretion system that injects effector molecules into gastric cells and subverts host cell signalling is associated with virulence. In this review we discuss the interplay of H. pylori colonization and virulence factors with host and environmental factors to determine disease outcome in infected individuals.
Collapse
Affiliation(s)
- Marguerite Clyne
- School of Medicine, University College Dublin, Dublin, Ireland
- The Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Tadhg Ó Cróinín
- The Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
3
|
Zhao J, Wang B, Yan M, Liu Y, Zhao R, Wang X, Shao T, Li Y, Imran M, Ji M, Zhao H, Guimarães CF, Li G, Zhou Q, Reis RL. Progress in enzyme-powered micro/nanomotors in diagnostics and therapeutics. Bioact Mater 2025; 46:555-568. [PMID: 39896991 PMCID: PMC11782855 DOI: 10.1016/j.bioactmat.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/21/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Enzyme-powered micro/nanomotors (EMNMs) represent cutting-edge research taking advantage of enzymes as biocatalysts to provide a driving force for micro/nanomotors. Up to now, EMNMs have been designed to be powered by catalase, urease, lipase, collagenase, compound enzymes, etc. They not only have good biocompatibility and biosafety but also possess the unique ability to utilize physiologically relevant fuel to achieve autonomous propulsion through in vivo catalytic reactions. This innovation has opened exciting possibilities for medical applications of EMNMs. Given the fact that the human body is naturally abundant with substrates available for enzymatic reactions, EMNMs can effectively exploit the complex microenvironment associated with diseases, enabling the diagnosis and treatment of various medical conditions. In this review, we first introduce different kinds of EMNMs applied in specific environments for the diagnosis and treatment of diseases, while highlighting their advancements for revolutionizing healthcare practices. Then, we address the challenges faced in this rapidly evolving field, and at last, the potential future development directions are discussed. As the potential of EMNMs becomes increasingly evident, continued research and exploration are essential to unlock their full capabilities and to ensure their successful integration into clinical applications.
Collapse
Affiliation(s)
- Jinpeng Zhao
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266113, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Banghui Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Mingzhe Yan
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Yuxin Liu
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Ruizhe Zhao
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Xuezhe Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Tianyi Shao
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Yifei Li
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Muhammad Imran
- Department of Biosciences, COMSATS University, Islamabad Park Road, Islamabad, Pakistan
| | - Mingze Ji
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Hong Zhao
- Department of Bone, Huangdao District Central Hospital, Qingdao, 266555, China
| | - Carlos F. Guimarães
- 3B's Research Group-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, 4805-017, Portugal
- ICVS/3B's – Portuguese Government Associate Laboratory, University of Minho, Braga, Guimarães, Portugal
| | - Guotai Li
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266113, China
| | - Qihui Zhou
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266113, China
| | - Rui L. Reis
- 3B's Research Group-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, 4805-017, Portugal
- ICVS/3B's – Portuguese Government Associate Laboratory, University of Minho, Braga, Guimarães, Portugal
| |
Collapse
|
4
|
Mandle HB, Jenab M, Gunter MJ, Tjønneland A, Olsen A, Dahm CC, Zhang J, Sugier PE, Rothwell J, Severi G, Kaaks R, Katzke VA, Schulze MB, Masala G, Sieri S, Panico S, Sacerdote C, Bonet C, Sánchez MJ, Amiano P, Huerta JM, Guevara M, Palmqvist R, Löwenmark T, Perez-Cornago A, Weiderpass E, Heath AK, Cross AJ, Vineis P, Hughes DJ, Fedirko V. Inflammation and gut barrier function-related genes and colorectal cancer risk in western European populations. Mutagenesis 2025; 40:48-60. [PMID: 38441165 PMCID: PMC11911009 DOI: 10.1093/mutage/geae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/04/2024] [Indexed: 03/21/2024] Open
Abstract
Gut barrier dysfunction and related inflammation are known to be associated with the development and progression of colorectal cancer (CRC). We investigated associations of 292 single-nucleotide polymorphisms (SNPs) from 27 genes related to endotoxins/lipopolysaccharide (LPS) sensing and tolerance, mucin synthesis, inflammation, and Crohn's disease with colon and rectal cancer risks. Incident CRC cases (N = 1374; colon = 871, rectum = 503) were matched 1:1 to controls nested within the European Prospective Investigation into Cancer and Nutrition cohort. Previously measured serum concentrations of gut barrier function and inflammation biomarkers (flagellin/LPS-specific immunoglobulins and C-reactive protein [CRP]) were available for a sub-set of participants (Ncases = 1001; Ncontrols = 667). Forty-two unique SNPs from 19 different genes were associated with serum biomarkers at Punadjusted ≤ 0.05 among controls. Among SNPs associated with a gut permeability score, 24 SNPs were in genes related to LPS sensing and mucin synthesis. Nine out of 12 SNPs associated with CRP were in genes related to inflammation or Crohn's disease. TLR4 was associated with colon cancer at the SNP level (nine SNPs, all Punadjusted ≤ 0.04) and at the gene level (Punadjusted ≤ 0.01). TLR4 rs10759934 was associated with rectal cancer but not colon cancer. Similarly, IL10 was associated with rectal cancer risk at an SNP and gene level (both Punadjusted ≤ 0.01), but not colon cancer. Genes and SNPs were selected a priori; therefore, we present unadjusted P-values. However, no association was statistically significant after multiple testing correction. This large and comprehensive study has identified gut barrier function and inflammation-related genes possibly contributing to CRC risk in European populations and is consistent with potential etiological links between host genetic background, gut barrier permeability, microbial endotoxemia, and CRC development.
Collapse
Affiliation(s)
- Hannah B Mandle
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Anne Tjønneland
- Diet, Cancer and Health, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
| | - Anja Olsen
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christina C Dahm
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jie Zhang
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Pierre-Emmanuel Sugier
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
- Laboratoire de Mathématiques et de leurs Applications de Pau E2S UPPA, CNRS, 64013 Pau Cedex, France
| | - Joseph Rothwell
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, 14469 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14469 Nuthetal, Germany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica E Chirurgia, Federico II University, 80131 Naples, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, AOU Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Catalina Bonet
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.18011 Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18071 Granada, Spain
| | - Pilar Amiano
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, BioGipuzkoa Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, 20014 Donostia – San Sebastian, Spain
| | - José María Huerta
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council-IMIB, 30120, El Palmar, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, 31003 Pamplona, Navarra, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Richard Palmqvist
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Thyra Löwenmark
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer, 69366 Lyon Cedex 07, France
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Paolo Vineis
- MRC Centre for Environment and Health, School of public Health, Imperial College London, London W2 1PG, UK
- Italian Institute for Genomic Medicine (IIGM), 10060 Candiolo TO,Italy
| | - David J Hughes
- Cancer Biology and Therapeutics Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Veronika Fedirko
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
- Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, 77030 Houston, TX, USA
| |
Collapse
|
5
|
Li WW, Yu ZL, Jia J. Urease-powered micro/nanomotors: Current progress and challenges. J Pharm Anal 2025; 15:101095. [PMID: 40177066 PMCID: PMC11964642 DOI: 10.1016/j.jpha.2024.101095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/26/2024] [Accepted: 09/02/2024] [Indexed: 04/05/2025] Open
Abstract
Enzyme-powered micro/nanomotors (MNMs) (EMNMs) use natural enzymes to facilitate the decomposition of fuels, including hydrogen peroxide (H2O2), glucose, triglycerides, and urea to provide power. EMNMs can achieve self-propulsion through the in situ utilization of biofuels without additional fuels, exhibiting excellent biocompatibility and significant potential for application in the biomedical field. Compared with H2O2, which may cause oxidative damage to the body, urea exhibits superior biosafety characteristics. Presently, urease-powered MNMs (UMNMs) have made notable progress in their applications in the biomedical field and have garnered considerable attention from researchers. In this review, we present the latest advancements in the biomedical field of UMNMs, primarily focusing on: 1) diverse materials used for constructing the fundamental framework of motors; 2) control of motor movement through the regulation of enzymatic reaction rates; and 3) research directions for the clinical application of motors, including in vivo imaging, biomarker detection, cancer treatment, optical therapy, overcoming biological barriers, antibacterial interventions, antithrombotic strategies, and gastric disease management. Despite showing immense potential in biomedical applications, there are still several challenges impeding its practical implementation, such as maintaining activity in the in vivo environment while accurately targeting specific sites to achieve the desired clinical therapeutic effects.
Collapse
Affiliation(s)
- Wen-Wen Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
6
|
Wang B, Luan J, Zhao W, Yu J, Li A, Li X, Zhong X, Cao H, Wang R, Liu B, Lu S, Shi M. Comprehensive multiomics analysis of the signatures of gastric mucosal bacteria and plasma metabolites across different stomach microhabitats in the development of gastric cancer. Cell Oncol (Dordr) 2025; 48:139-159. [PMID: 38963518 PMCID: PMC11850404 DOI: 10.1007/s13402-024-00965-3] [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] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE As an important component of the microenvironment, the gastric microbiota and its metabolites are associated with tumour occurrence, progression, and metastasis. However, the relationship between the gastric microbiota and the development of gastric cancer is unclear. The present study investigated the role of the gastric mucosa microbiome and metabolites as aetiological factors in gastric carcinogenesis. METHODS Gastric biopsies from different stomach microhabitats (n = 70) were subjected to 16S rRNA gene sequencing, and blood samples (n = 95) were subjected to untargeted metabolome (gas chromatography‒mass spectrometry, GC‒MS) analyses. The datasets were analysed using various bioinformatics approaches. RESULTS The microbiota diversity and community composition markedly changed during gastric carcinogenesis. High Helicobacter. pylori colonization modified the overall diversity and composition of the microbiota associated with gastritis and cancer in the stomach. Most importantly, analysis of the functional features of the microbiota revealed that nitrate reductase genes were significantly enriched in the tumoral microbiota, while urease-producing genes were significantly enriched in the microbiota of H. pylori-positive patients. A panel of 81 metabolites was constructed to discriminate gastric cancer patients from gastritis patients, and a panel of 15 metabolites was constructed to discriminate H. pylori-positive patients from H. pylori-negative patients. receiver operator characteristic (ROC) curve analysis identified a series of gastric microbes and plasma metabolites as potential biomarkers of gastric cancer. CONCLUSION The present study identified a series of signatures that may play important roles in gastric carcinogenesis and have the potential to be used as biomarkers for diagnosis and for the surveillance of gastric cancer patients with minimal invasiveness.
Collapse
Affiliation(s)
- Bingsen Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Jiahui Luan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
| | - Weidong Zhao
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Junbao Yu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Anqing Li
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Xinxin Li
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Xiaoqin Zhong
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Hongyun Cao
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
| | - Ruicai Wang
- Department of Pathology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Bo Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
- Department of Pulmonary and Critical Care Medicine, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Shiyong Lu
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Mei Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China.
| |
Collapse
|
7
|
Duan Y, Xu Y, Dou Y, Xu D. Helicobacter pylori and gastric cancer: mechanisms and new perspectives. J Hematol Oncol 2025; 18:10. [PMID: 39849657 PMCID: PMC11756206 DOI: 10.1186/s13045-024-01654-2] [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: 08/20/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer remains a significant global health challenge, with Helicobacter pylori (H. pylori) recognized as a major etiological agent, affecting an estimated 50% of the world's population. There has been a rapidly expanding knowledge of the molecular and pathogenetic mechanisms of H. pylori over the decades. This review summarizes the latest research advances to elucidate the molecular mechanisms underlying the H. pylori infection in gastric carcinogenesis. Our investigation of the molecular mechanisms reveals a complex network involving STAT3, NF-κB, Hippo, and Wnt/β-catenin pathways, which are dysregulated in gastric cancer caused by H. pylori. Furthermore, we highlight the role of H. pylori in inducing oxidative stress, DNA damage, chronic inflammation, and cell apoptosis-key cellular events that pave the way for carcinogenesis. Emerging evidence also suggests the effect of H. pylori on the tumor microenvironment and its possible implications for cancer immunotherapy. This review synthesizes the current knowledge and identifies gaps that warrant further investigation. Despite the progress in our previous knowledge of the development in H. pylori-induced gastric cancer, a comprehensive investigation of H. pylori's role in gastric cancer is crucial for the advancement of prevention and treatment strategies. By elucidating these mechanisms, we aim to provide a more in-depth insights for the study and prevention of H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Yantao Duan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yonghu Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Dou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Wang S, Mu Y, Zhang J, Wang C. Prognostic and clinicopathological significance of mucin family members expression in gastric cancer: a meta-analysis. Front Oncol 2025; 14:1512971. [PMID: 39886661 PMCID: PMC11779608 DOI: 10.3389/fonc.2024.1512971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/13/2024] [Indexed: 02/01/2025] Open
Abstract
Background Mucin family members have been reported to be widely expressed in gastric carcinoma with diverse functions. Several important mucins exert the function of tumorigenesis or progression in gastric cancer (GC). Here, we conduct this meta-analysis to evaluate the association between mucin expression and clinicopathological features in GC. Methods Literature searches were performed in PubMed, Embase, The Cochrane Library, and ISI Web of Science, and, finally, 28 studies met our criteria. Odds ratios or hazard ratios with 95% confidence intervals were calculated to evaluate the effect quantity. We analyzed the expression of MUC1, MUC2, MUC5AC, and MUC6 and their clinicopathological characteristics separately at the same time. Results Twenty-eight studies that contain 4,603 patients were included in our meta-analysis. MUC1 was associated with gender, Lauren classification, depth of tumor invasion, TNM, vascular invasion, lymph metastasis, and lymphatic invasion, WHO grade, as well as the 5-year survival rate. MUC2 was significantly correlated with lymphatic invasion and WHO grade. MUC5AC was highly positive in gender, depth of tumor invasion, WHO grade, TNM, lymph metastasis, and lymphatic invasion. Moreover, cases with decreased MUC5AC expression were correlated with less 5-year survival. MUC6 was only related with lymphatic invasion. Conclusion Our meta-analysis showed that MUC1 and MUC5AC had prognostic value in GC detected by immunohistochemistry. MUC1 and MUC5AC were also associated with some other significant clinicopathological parameters. Moreover, MUC2 and MUC6 also exert their influence in lymphatic invasion. However, further enlarged study awaits to verify our conclusion to deeply explore the role of mucin family members in GC.
Collapse
Affiliation(s)
| | | | | | - Chengfeng Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
McCoy R, Wang K, Treiber J, Fu Y, Malliaras GG, Salleo A, Owens RM. Mucus-on-a-chip: investigating the barrier properties of mucus with organic bioelectronics. J Mater Chem B 2025; 13:577-587. [PMID: 39575664 DOI: 10.1039/d4tb01351d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Gastrointestinal (GI) mucus is a biologically complex hydrogel that acts as a partially permeable barrier between the contents of the GI tract and the mucosal epithelial lining. Its structural integrity is essential for the lubrication of the tract thereby aiding smooth transit of contents, and the protection of the epithelium from pathogens that seek to colonise and invade. Understanding its physical response to drugs and the microbiome is essential for treating many gastrointestinal infectious diseases. Given this, a static in vitro model of a GI mucus-on-a-chip has been developed with integrated electronics to monitor the barrier properties of mucus hydrogels. Its application for investigating the effect of drugs and biofilm formation on the mucus structure is validated using rheological techniques, confocal microscopy and electrochemical impedance spectroscopy (EIS).
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
- Department of Electrical Engineering, University of Cambridge, CB3 0FA, Cambridge, UK
| | - Kaixin Wang
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Jeremy Treiber
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Ying Fu
- Department of Pure and Applied Chemistry, University of Strathclyde, G1 1XQ, Glasgow, UK
| | - George G Malliaras
- Department of Electrical Engineering, University of Cambridge, CB3 0FA, Cambridge, UK
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| |
Collapse
|
10
|
Chen N, Yao P, Farid MS, Zhang T, Luo Y, Zhao C. Effect of bioactive compounds in processed Camellia sinensis tea on the intestinal barrier. Food Res Int 2025; 199:115383. [PMID: 39658174 DOI: 10.1016/j.foodres.2024.115383] [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: 08/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
The human intestinal tract plays a pivotal role in safeguarding the body against noxious substances and microbial pathogens by functioning as a barrier. This barrier function is achieved through the combined action of physical, chemical, microbial, and immune components. Tea (Camellia sinensis) is the most widely consumed beverage in the world, and it is consumed and appreciated in a multitude of regions across the globe. Tea can be classified into various categories, including green, white, yellow, oolong, black, and dark teas, based on the specific processing methods employed. In recent times, there has been a notable surge in scientific investigation into the various types of tea. The recent surge in research on tea can be attributed to the plethora of bioactive compounds it contains, including polyphenols, polysaccharides, pigments, and theanine. The processing of different teas affects the active ingredients to varying degrees, resulting in a range of chemical reactions and the formation of different types and quantities of ingredients. The bioactive compounds present in tea are of great importance for the maintenance of the integrity of the intestinal barrier, operating through a variety of mechanisms. This literature review synthesizes scientific studies on the impact of the primary bioactive compounds and different processing methods of tea on the intestinal barrier function. This review places particular emphasis on the exploration of the barrier repair and regulatory effects of these compounds, including the mitigation of damage to different barriers following intestinal diseases. Specifically, the active ingredients in tea can alleviate damage to physical barriers and chemical barriers by regulating barrier protein expression. At the same time, they can also maintain the stability of immune and biological barriers by regulating the expression of inflammatory factors and the metabolism of intestinal flora. This investigation can establish a strong theoretical foundation for the future development of innovative tea products.
Collapse
Affiliation(s)
- Nan Chen
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Peng Yao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | | | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Yangchao Luo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, United States.
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
11
|
Rosinke K, Tachiyama S, Mrásek J, Liu J, Hoover TR. A Helicobacter pylori flagellar motor accessory is needed to maintain the barrier function of the outer membrane during flagellar rotation. PLoS Pathog 2025; 21:e1012860. [PMID: 39792952 PMCID: PMC11756786 DOI: 10.1371/journal.ppat.1012860] [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: 03/11/2024] [Revised: 01/23/2025] [Accepted: 12/26/2024] [Indexed: 01/12/2025] Open
Abstract
The Helicobacter pylori flagellar motor contains several accessory structures that are not found in the archetypal Escherichia coli and Salmonella enterica motors. H. pylori hp0838 encodes a previously uncharacterized lipoprotein and is in an operon with flgP, which encodes a motor accessory protein. Deletion analysis of hp0838 in H. pylori B128 showed that the gene is not required for motility in soft agar medium, but the mutant displayed a reduced growth rate and an increased sensitivity to bacitracin, which is an antibiotic that is normally excluded by the outer membrane. Introducing a plasmid-borne copy of hp0838 into the H. pylori Δhp0838 mutant suppressed the fitness defect and antibiotic sensitivity of the strain. A variant of the Δhp0838 mutant containing a frameshift mutation in pflA, which resulted in paralyzed flagella, displayed wild-type growth rate and resistance to bacitracin, suggesting the fitness defect and antibiotic sensitivity of the Δhp0838 mutant are dependent on flagellar rotation. Comparative analysis of in-situ structures of the wild type and Δhp0838 mutant motors revealed the Δhp0838 mutant motor lacked a previously undescribed ring structure with 18-fold symmetry located near the outer membrane. Given its role in formation of the motor outer ring, HP0838 was designated FapH (flagellar accessory protein in Helicobacter pylori) and the motor accessory formed the protein was named the FapH ring. Our data suggest that the FapH ring helps to preserve outer membrane barrier function during flagellar rotation. Given that FapH homologs are present in many members of the phylum Campylobacterota, they may have similar roles in protecting the outer membrane from damage due to flagellar rotation in these bacteria.
Collapse
Affiliation(s)
- Kyle Rosinke
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| | - Shoichi Tachiyama
- Department of Microbial Pathogenesis, Microbial Sciences Institute, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Jan Mrásek
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
| | - Jun Liu
- Department of Microbial Pathogenesis, Microbial Sciences Institute, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Timothy R. Hoover
- Department of Microbiology, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
12
|
Properzi S, Stracci F, Rosi M, Lupi C, Villarini A, Gili A. Can a diet rich in Brassicaceae help control Helicobacter pylori infection? A systematic review. Front Med (Lausanne) 2024; 11:1454902. [PMID: 39741515 PMCID: PMC11685009 DOI: 10.3389/fmed.2024.1454902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/11/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Helicobacter pylori (Hp) infection is highly prevalent globally and poses a significant public health challenge due to its link with chronic gastritis, peptic ulcers, and gastric malignancies. Hp's persistence within the gastric environment, particularly in case of infection with virulent strains, triggers chronic inflammatory responses and mucosal damage. Antibiotic therapy is the primary approach for Hp eradication, but antibiotic resistance and adverse effects hinder treatment efficacy. Emerging evidence suggests that Brassicaceae-derived metabolites could serve as adjunctive therapy for Hp infection, offering potential antimicrobial and anti-inflammatory benefits. Methods A systematic literature review was conducted following PRISMA guidelines to assess the impact of Brassicaceae-rich diets on Hp infection control. Searches were performed in MEDLINE PubMed, Web of Science, and the Cochrane Library until 18 October 2023, without language or date restrictions. Eligible studies meeting PICOS criteria were included, encompassing populations infected with Hp or Hp-infected human cell cultures, interventions involving Brassicaceae consumption or its bioactive molecules, and outcomes related to Hp infection control, antibiotic therapy interactions, reduction of antibiotic side effects, and inflammation mitigation. Animal studies, cell line experiments, reviews unrelated to the research objectives, and studies on Hp-related gastric cancer were excluded. Results Available evidence indicates that Brassicaceae consumption exhibits the potential to reduce Hp colonization but achieving complete eradication of the pathogen remains challenging. Conflicting results regarding the efficacy of broccoli in Hp treatment emerge, with certain investigations suggesting limited effectiveness. Other studies point to a potential for heightened eradication rates when combined with standard triple therapy. Furthermore, promising outcomes are observed with broccoli extract supplements, indicating their role in mitigating Hp-induced gastric mucosal damage. In fact, it is noteworthy that sulforaphane and its derivatives manifest notable reductions in pro-inflammatory markers, indicative of their anti-inflammatory properties. Adverse events associated with antibiotic therapy seem unaffected by sulforaphane derivatives or probiotics. However, individual responses to these treatments vary, underscoring the unpredictability of their efficacy in ameliorating antibiotic therapy-related side effects. Conclusion Our systematic review highlights the potential of Brassicaceae-rich diets as adjunctive therapy for Hp infection, offering synergistic interactions with antibiotics and possibly mitigating antibiotic side effects and inflammation. Further research, particularly well-designed randomized trials, is warranted to elucidate the therapeutic efficacy and optimal utilization of Brassicaceae-derived metabolites in managing human Hp-related diseases.
Collapse
Affiliation(s)
- Sara Properzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Fabrizio Stracci
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Margherita Rosi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Chiara Lupi
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Anna Villarini
- Department of Medicine and Surgery, University of Perugia, Perugia, Umbria, Italy
| | - Alessio Gili
- Department of Life Sciences, Health and Health Professions, Link Campus University, Rome, Italy
| |
Collapse
|
13
|
Hazt B, Read DJ, Harlen OG, Poon WCK, O'Connell A, Sarkar A. Mucoadhesion across scales: Towards the design of protein-based adhesives. Adv Colloid Interface Sci 2024; 334:103322. [PMID: 39489118 DOI: 10.1016/j.cis.2024.103322] [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: 03/26/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Mucoadhesion is a special case of bioadhesion in which a material adheres to soft mucosal tissues. This review elucidates our current understanding of mucoadhesion across length, time, and energy scales by focusing on relevant structural features of mucus. We highlight the importance of both covalent and non-covalent interactions that can be tailored to maximize mucoadhesive interactions, particularly concerning proteinaceous mucoadhesives, which have been explored only to a limited extent so far in the literature. In particular, we highlight the importance of thiol groups, hydrophobic moieties, and charged species inherent to proteins as key levers to fine tune mucoadhesive performance. Some aspects of protein surface modification by grafting specific functional groups or coupling with polysaccharides to influence mucoadhesive performance are examined. Insights from this review offer a physicochemical roadmap to inform the development of biocompatible, protein-based mucoadhesive systems that can fulfil dual roles for both adhesion and delivery of actives, enabling the fabrication of advanced biomedical, nutritional and allied soft material technologies.
Collapse
Affiliation(s)
- Bianca Hazt
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK
| | - Daniel J Read
- School of Mathematics, University of Leeds, LS2 9JT, UK
| | | | - Wilson C K Poon
- School of Physics and Astronomy, University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - Adam O'Connell
- Polymer Science Platform, Reckitt Benckiser Healthcare (UK) Ltd, Dansom Lane S, Hull, HU8 7DS, UK
| | - Anwesha Sarkar
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
14
|
Miller MA, Medina S. Synthetic Colonic Mucus Enables the Development of Modular Microbiome Organoids. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2402514. [PMID: 39309137 PMCID: PMC11415244 DOI: 10.1002/adfm.202402514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Indexed: 09/25/2024]
Abstract
The human colon is home to trillions of microorganisms that modulate gastrointestinal physiology. Our understanding of how this gut ecosystem impacts human health, although evolving, has been slowed by the lack of accessible tools suitable to studying complex host-mucus-microbe interactions. Here, we report a synthetic gel-like material capable of recapitulating the varied structural, mechanical, and biochemical profiles of native human colonic mucus to develop compositionally simple microbiome screening platforms with utility in microbiology and drug discovery. The viscous fibrillar material is realized through templated assembly of a fluorine-rich amino acid at liquid-liquid interphases. The fluorine-assisted mucus surrogate (FAMS) can be decorated with mucins to serve as a habitat for microbial colonization and integrated with human colorectal cells to generate artificial mucosae, referred to as a microbiome organoid. Notably, FAMS are made with inexpensive and commercially available materials, and can be generated using simple protocols and standard laboratory hardware. As a result, this platform can be broadly incorporated into various laboratory settings to advance probiotic research and inform in vivo approaches. If implemented into high throughput screening approaches, FAMS may represent a valuable tool to study compound metabolism and gut permeability, with an exemplary demonstration of this utility presented here.
Collapse
Affiliation(s)
- Michael A Miller
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA, 16802-4400
| | - Scott Medina
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA, 16802-4400
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA, 16802-4400
| |
Collapse
|
15
|
Krzyżek P, Migdał P, Krzyżanowska B, Duda-Madej A. Optimization of Helicobacter pylori Biofilm Formation in In Vitro Conditions Mimicking Stomach. Int J Mol Sci 2024; 25:9839. [PMID: 39337326 PMCID: PMC11432336 DOI: 10.3390/ijms25189839] [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: 08/13/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Helicobacter pylori is one of the most common bacterial pathogens worldwide and the main etiological agent of numerous gastric diseases. The frequency of multidrug resistance of H. pylori is growing and the leading factor related to this phenomenon is its ability to form biofilm. Therefore, the establishment of a proper model to study this structure is of critical need. In response to this, the aim of this original article is to validate conditions of the optimal biofilm development of H. pylori in monoculture and co-culture with a gastric cell line in media simulating human fluids. Using a set of culture-based and microscopic techniques, we proved that simulated transcellular fluid and simulated gastric fluid, when applied in appropriate concentrations, stimulate autoaggregation and biofilm formation of H. pylori. Additionally, using a co-culture system on semi-permeable membranes in media imitating the stomach environment, we were able to obtain a monolayer of a gastric cell line with H. pylori biofilm on its surface. We believe that the current model for H. pylori biofilm formation in monoculture and co-culture with gastric cells in media containing host-mimicking fluids will constitute a platform for the intensification of research on H. pylori biofilms in in vitro conditions that simulate the human body.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (B.K.); (A.D.-M.)
| | - Paweł Migdał
- Department of Bees Breeding, Institute of Animal Husbandry, Wroclaw University of Environmental and Life Sciences, 51-630 Wroclaw, Poland;
| | - Barbara Krzyżanowska
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (B.K.); (A.D.-M.)
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (B.K.); (A.D.-M.)
| |
Collapse
|
16
|
Shirani M, Shariati S, Bazdar M, Sojoudi Ghamnak F, Moradi M, Shams Khozani R, Taki E, Arabsorkhi Z, Heidary M, Eskandari DB. The immunopathogenesis of Helicobacter pylori-induced gastric cancer: a narrative review. Front Microbiol 2024; 15:1395403. [PMID: 39035439 PMCID: PMC11258019 DOI: 10.3389/fmicb.2024.1395403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Helicobacter pylori infection is a well-established risk factor for the development of gastric cancer (GC). Understanding the immunopathogenesis underlying this association is crucial for developing effective preventive and therapeutic strategies. This narrative review comprehensively explores the immunopathogenesis of H. pylori-induced GC by delving into several key aspects, emphasizing the pivotal roles played by H. pylori virulence factors, including cytotoxin-associated gene A (cagA) and vacuolating cytotoxin A (vacA), blood group antigen-binding adhesin (babA), and sialic acid binding adhesin (sabA). Moreover, the review focuses on the role of toll-like receptors (TLRs) and cytokines in the complex interplay between chronic infection and gastric carcinogenesis. Finally, the study examines the association between H. pylori evasion of the innate and adaptive immune response and development of GC. A comprehensive understanding of the immunopathogenesis of H. pylori-induced GC is essential for designing targeted interventions to prevent and manage this disease. Further research is warranted to elucidate the intricate immune responses involved and identify potential therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Maryam Shirani
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeedeh Shariati
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Monireh Bazdar
- School of Medicine, Razi Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Melika Moradi
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Arabsorkhi
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | |
Collapse
|
17
|
Wan Y, Xu Z, Zhu S, Zhou Y, Lü X, Shan Y. Dynamic changes in the aggregation-depolymerization behavior of Ovomucin-Complex and its binding to urease during in vitro simulated gastric digestion. Int J Biol Macromol 2024; 270:132295. [PMID: 38735615 DOI: 10.1016/j.ijbiomac.2024.132295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Ovomucin-Complex extracted from egg white is expected to have a barrier function similar to gastric mucin. In this study, the dynamic changes in structure, rheological properties and binding ability of Ovomucin-Complex during in vitro simulated gastric digestion were investigated. The results from HPLC and CLSM showed that extremely acidic pH (pH = 2.0) promoted Ovomucin-Complex to form aggregation. Acid-induced aggregation may hinder its binding to pepsin, thus rendering Ovomucin-Complex resistant to pepsin. Consequently, most of the polymer structure and weak gel properties of Ovomucin-Complex retained after simulated gastric digestion as verified by HPLC, CLSM and rheological measurement, although there was a small breakdown of the glycosidic bond as confirmed by the increased content of reducing sugar. The significantly reduced hydrophobic interactions of Ovomucin-Complex were observed under extremely acidic conditions and simulated gastric digestion compared with the native. Noticeably, the undigested Ovomucin-Complex after simulated gastric digestion showed a higher affinity (KD = 5.0 ± 3.2 nm) for urease - the key surface antigen of Helicobacter pylori. The interaction mechanism between Ovomucin-Complex and urease during gastric digestion deserves further studies. This finding provides a new insight to develop an artificial physical mucus barrier to reduce Helicobacter pylori infection.
Collapse
Affiliation(s)
- Yanqing Wan
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Zhiman Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Shengnan Zhu
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Yuan Zhou
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Yuanyuan Shan
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China.
| |
Collapse
|
18
|
Le Roy H, Song J, Lundberg D, Zhukhovitskiy AV, Johnson JA, McKinley GH, Holten-Andersen N, Lenz M. Valence can control the nonexponential viscoelastic relaxation of multivalent reversible gels. SCIENCE ADVANCES 2024; 10:eadl5056. [PMID: 38748785 PMCID: PMC11095449 DOI: 10.1126/sciadv.adl5056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Gels made of telechelic polymers connected by reversible cross-linkers are a versatile design platform for biocompatible viscoelastic materials. Their linear response to a step strain displays a fast, near-exponential relaxation when using low-valence cross-linkers, while larger supramolecular cross-linkers bring about much slower dynamics involving a wide distribution of timescales whose physical origin is still debated. Here, we propose a model where the relaxation of polymer gels in the dilute regime originates from elementary events in which the bonds connecting two neighboring cross-linkers all disconnect. Larger cross-linkers allow for a greater average number of bonds connecting them but also generate more heterogeneity. We characterize the resulting distribution of relaxation timescales analytically and accurately reproduce stress relaxation measurements on metal-coordinated hydrogels with a variety of cross-linker sizes including ions, metal-organic cages, and nanoparticles. Our approach is simple enough to be extended to any cross-linker size and could thus be harnessed for the rational design of complex viscoelastic materials.
Collapse
Affiliation(s)
- Hugo Le Roy
- Université Paris-Saclay, CNRS, LPTMS, 91405, Orsay, France
- Institute of Physics, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Jake Song
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - David Lundberg
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Aleksandr V. Zhukhovitskiy
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Gareth H. McKinley
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Niels Holten-Andersen
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Bioengineering and Materials Science and Engineering, Lehigh University, Bethlehem, PA 18015, USA
| | - Martin Lenz
- Université Paris-Saclay, CNRS, LPTMS, 91405, Orsay, France
- PMMH, CNRS, ESPCI Paris, PSL University, Sorbonne Université, Université de Paris, F-75005 Paris, France
| |
Collapse
|
19
|
Ethier R, Krishnamurthy A, Jeffrey M, Tompkins TA. Profiling of Metabolites in a Fermented Soy Dietary Supplement Reinforces its Role in the Management of Intestinal Inflammation. Mol Nutr Food Res 2024; 68:e2300770. [PMID: 38522032 DOI: 10.1002/mnfr.202300770] [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: 10/31/2023] [Revised: 02/19/2024] [Indexed: 03/25/2024]
Abstract
SCOPE Gastro-AD (GAD) is a soy flour derived product that undergoes an industrial fermentation with Lactobacillus delbrueckii R0187 and has demonstrated clinical effects in gastroesophageal reflux and peptic ulcer symptom resolution. The aim of this study is to describe and link GAD's metabolomic profile to plausible mechanisms that manifest and explain the documented clinical outcomes. METHODS AND RESULTS 1H NMR spectroscopy with multivariate statistical analysis is used to characterize the prefermented soy flour and GAD products. The acquired spectra are screened using various resources and the molecular assignments are confirmed using total correlation spectroscopy (TOCSY). Peaks corresponding to different metabolites are integrated and compared between the two products for relative changes. HPLC and GC are used to quantify some specific molecules. NMR analyses demonstrate significant changes in the composition of various assigned bioactive moieties. HPLC and GC analysis demonstrate deglycation of isoflavones after fermentation, resulting in estrogenically active secondary metabolites that have been previously shown to help to reduce inflammation. CONCLUSION The identification of bioactive molecules, such as genistein and SCFAs, capable of modulating anti-inflammatory signaling cascades in the stomach's gastric and neuroendocrine tissues can explain the reported biological effects in GAD and is supported by in vivo data.
Collapse
Affiliation(s)
- Richard Ethier
- Richard Ethier Consulting, Montreal, Quebec, H4C 2J9, Canada
| | - Arun Krishnamurthy
- Purity-IQ Inc., Suite# 102, 150 Research Lane, Guelph, Ontario, N1G 4T2, Canada
| | - Michael Jeffrey
- Faculty of Science, Engineering & Information Technology, Durham College, Oshawa, Ontario, L1G 0C5, Canada
| | - Thomas A Tompkins
- Lallemand Bio-Ingredients, 1620 rue Prefontaine, Montreal, Quebec, H1W 2N8, Canada
| |
Collapse
|
20
|
Jiang Y, Mi L, Xu X, Hii ARK, Wu Z, Qi X. Urease catalyzed high-density sodium alginate microspheres enable high oral bioavailability of macromolecular drugs. Biomater Sci 2024; 12:1515-1528. [PMID: 38284628 DOI: 10.1039/d3bm01715j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Destruction of insulin caused by the gastric microenvironment and rapid deactivation pose inevitable barriers to oral macromolecular absorption, especially for most peptide and protein drugs. In this study, we developed high-density sodium alginate microspheres composed of magnesium oxide and urease to address these challenges. These microspheres aim to anchor the gastric mucus layer and induce microenvironmental liquefaction, thereby enhancing gastric retention and the protection of insulin. The sedimentation test confirmed the capability of the Ins/Ur/MgO@SA microsphere to rapidly traverse the gastric juice under the influence of gravity. Additionally, the urease immobilized on the Ins/Ur/MgO@SA microspheres catalyzes the hydrolysis of urea in the gastric mucus and promotes the liquefaction of mucus, which is beneficial for microsphere retention. The inclusion of MgO particles and urease, acting as pHM modifiers, helps in adjusting the local pH to avoid gastric acid-induced damage. Subsequently, an in vivo pharmacokinetic experiment verified that the relative bioavailability of the p.o. Ins/Ur/MgO@SA treated group was 15-fold higher than that of the p.o.insulin treated group. Meanwhile, satisfactory blood glucose level (BGL) reduction was observed in diabetic animals. In conclusion, Ins/Ur/MgO@SA microspheres demonstrate high biocompatibility as insulin carriers with prolonged drug release time and increased gastric retention properties, showing a far-reaching strategy for oral macromolecular drug delivery.
Collapse
Affiliation(s)
- Yicheng Jiang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Li Mi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Xiang Xu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
- King's College London, Institution of Pharmaceutical Science, Franklin Wilkins Building, 150 Stamford St, London SE1 9NH, England, UK
| | - Adric Ru Khiing Hii
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
- Industrial Technology Innovation Platform, Zhejiang Center for Safety Study of Drug Substances, Hangzhou 310018, China
| |
Collapse
|
21
|
Ruiz-González N, Esporrín-Ubieto D, Hortelao AC, Fraire JC, Bakenecker AC, Guri-Canals M, Cugat R, Carrillo JM, Garcia-Batlletbó M, Laiz P, Patiño T, Sánchez S. Swarms of Enzyme-Powered Nanomotors Enhance the Diffusion of Macromolecules in Viscous Media. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309387. [PMID: 38200672 DOI: 10.1002/smll.202309387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Indexed: 01/12/2024]
Abstract
Over the past decades, the development of nanoparticles (NPs) to increase the efficiency of clinical treatments has been subject of intense research. Yet, most NPs have been reported to possess low efficacy as their actuation is hindered by biological barriers. For instance, synovial fluid (SF) present in the joints is mainly composed of hyaluronic acid (HA). These viscous media pose a challenge for many applications in nanomedicine, as passive NPs tend to become trapped in complex networks, which reduces their ability to reach the target location. This problem can be addressed by using active NPs (nanomotors, NMs) that are self-propelled by enzymatic reactions, although the development of enzyme-powered NMs, capable of navigating these viscous environments, remains a considerable challenge. Here, the synergistic effects of two NMs troops, namely hyaluronidase NMs (HyaNMs, Troop 1) and urease NMs (UrNMs, Troop 2) are demonstrated. Troop 1 interacts with the SF by reducing its viscosity, thus allowing Troop 2 to swim more easily through the SF. Through their collective motion, Troop 2 increases the diffusion of macromolecules. These results pave the way for more widespread use of enzyme-powered NMs, e.g., for treating joint injuries and improving therapeutic effectiveness compared with traditional methods.
Collapse
Affiliation(s)
- Noelia Ruiz-González
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - David Esporrín-Ubieto
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ana C Hortelao
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Juan C Fraire
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Anna C Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Marta Guri-Canals
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ramón Cugat
- Mutualidad de Futbolistas - Delegación Catalana, Federación Española de Fútbol, Barcelona, 08010, Spain
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - José María Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain. García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, 46115, Spain
| | | | - Patricia Laiz
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - Tania Patiño
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, 5612 AZ, The Netherlands
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
- Institució Catalana de Recerca i Estudies Avancats (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
22
|
Garg A, Karhana S, Khan MA. Nanomedicine for the eradication of Helicobacter pylori: recent advances, challenges and future perspective. Future Microbiol 2024; 19:431-447. [PMID: 38381027 DOI: 10.2217/fmb-2023-0189] [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: 08/23/2023] [Accepted: 10/31/2023] [Indexed: 02/22/2024] Open
Abstract
Helicobacter pylori infection is linked to gastritis, ulcers and gastric cancer. Nanomedicine offers a promising solution by utilizing nanoparticles for precise drug delivery, countering antibiotic resistance and delivery issues. Nanocarriers such as liposomes and nanoparticles enhance drug stability and circulation, targeting infection sites through gastric mucosa characteristics. Challenges include biocompatibility, stability, scalability and personalized therapies. Despite obstacles, nanomedicine's potential for reshaping H. pylori eradication is significant and showcased in this review focusing on benefits, limitations and future prospects of nanomedicine-based strategies.
Collapse
Affiliation(s)
- Aakriti Garg
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
- Centre for Translational & Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Sonali Karhana
- Centre for Translational & Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd A Khan
- Centre for Translational & Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
23
|
Fan J, Zhu J, Xu H. Strategies of Helicobacter pylori in evading host innate and adaptive immunity: insights and prospects for therapeutic targeting. Front Cell Infect Microbiol 2024; 14:1342913. [PMID: 38469348 PMCID: PMC10925771 DOI: 10.3389/fcimb.2024.1342913] [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: 11/22/2023] [Accepted: 02/08/2024] [Indexed: 03/13/2024] Open
Abstract
Helicobacter pylori (H. pylori) is the predominant pathogen causing chronic gastric mucosal infections globally. During the period from 2011 to 2022, the global prevalence of H. pylori infection was estimated at 43.1%, while in China, it was slightly higher at approximately 44.2%. Persistent colonization by H. pylori can lead to gastritis, peptic ulcers, and malignancies such as mucosa-associated lymphoid tissue (MALT) lymphomas and gastric adenocarcinomas. Despite eliciting robust immune responses from the host, H. pylori thrives in the gastric mucosa by modulating host immunity, particularly by altering the functions of innate and adaptive immune cells, and dampening inflammatory responses adverse to its survival, posing challenges to clinical management. The interaction between H. pylori and host immune defenses is intricate, involving evasion of host recognition by modifying surface molecules, manipulating macrophage functionality, and modulating T cell responses to evade immune surveillance. This review analyzes the immunopathogenic and immune evasion mechanisms of H. pylori, underscoring the importance of identifying new therapeutic targets and developing effective treatment strategies, and discusses how the development of vaccines against H. pylori offers new hope for eradicating such infections.
Collapse
Affiliation(s)
- Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Keane DP, Constantine CJ, Mellor MD, Poling-Skutvik R. Nanoparticle transport in biomimetic polymer-linked emulsions. AIChE J 2024; 70:e18307. [PMID: 40017798 PMCID: PMC11867629 DOI: 10.1002/aic.18307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/31/2023] [Indexed: 03/01/2025]
Abstract
The ability of nanoparticles to penetrate and transport through soft tissues is essential to delivering therapeutics to treat diseases or signaling agents for advanced imaging and sensing. Nanoparticle transport in biological systems, however, is challenging to predict and control due to the physicochemical complexity of tissues and biological fluids. Here, we demonstrate that nanoparticles suspended in a novel class of soft matter-polymer-linked emulsions (PLEs)-exhibit characteristics essential for mimicking transport in biological systems, including subdiffusive dynamics, non-Gaussian displacement distributions, and decoupling of dynamics from material viscoelasticity. Using multiple particle tracking, we identify the physical mechanisms underlying this behavior, which we attribute to a coupling of nanoparticle dynamics to fluctuations in the local network of polymer-linked droplets. Our findings demonstrate the potential of PLEs to serve as fully synthetic mimics of biological transport.
Collapse
Affiliation(s)
- Daniel P Keane
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Colby J Constantine
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Matthew D Mellor
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Ryan Poling-Skutvik
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| |
Collapse
|
25
|
Hanio S, Möllmert S, Möckel C, Choudhury S, Höpfel AI, Zorn T, Endres S, Schlauersbach J, Scheller L, Keßler C, Scherf-Clavel O, Bellstedt P, Schubert US, Pöppler AC, Heinze KG, Guck J, Meinel L. Bile Is a Selective Elevator for Mucosal Mechanics and Transport. Mol Pharm 2023; 20:6151-6161. [PMID: 37906224 DOI: 10.1021/acs.molpharmaceut.3c00550] [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: 11/02/2023]
Abstract
Mucus mechanically protects the intestinal epithelium and impacts the absorption of drugs, with a largely unknown role for bile. We explored the impacts of bile on mucosal biomechanics and drug transport within mucus. Bile diffused with square-root-of-time kinetics and interplayed with mucus, leading to transient stiffening captured in Brillouin images and a concentration-dependent change from subdiffusive to Brownian-like diffusion kinetics within the mucus demonstrated by differential dynamic microscopy. Bile-interacting drugs, Fluphenazine and Perphenazine, diffused faster through mucus in the presence of bile, while Metoprolol, a drug with no bile interaction, displayed consistent diffusion. Our findings were corroborated by rat studies, where co-dosing of a bile acid sequestrant substantially reduced the bioavailability of Perphenazine but not Metoprolol. We clustered over 50 drugs based on their interactions with bile and mucin. Drugs that interacted with bile also interacted with mucin but not vice versa. This study detailed the dynamics of mucus biomechanics under bile exposure and linked the ability of a drug to interact with bile to its abbility to interact with mucus.
Collapse
Affiliation(s)
- Simon Hanio
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Susobhan Choudhury
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Andreas I Höpfel
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Theresa Zorn
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Sebastian Endres
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Jonas Schlauersbach
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Lena Scheller
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Christoph Keßler
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Peter Bellstedt
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Institute for Clinical Chemistry, University of Zürich,Rämistrasse 100, 8091 Zurich, Switzerland
| | - Ulrich S Schubert
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), University of Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ann-Christin Pöppler
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Josef-Schneider-Strasse 2, 97080 Wuerzburg, Germany
| |
Collapse
|
26
|
Governa P, Romagnoli G, Albanese P, Rossi F, Manetti F, Biagi M. Effect of in vitro simulated digestion on the anti- Helicobacter Pylori activity of different Propolis extracts. J Enzyme Inhib Med Chem 2023; 38:2183810. [PMID: 36916299 PMCID: PMC10026752 DOI: 10.1080/14756366.2023.2183810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Helicobacter pylori (HP) is among the most common pathogens causing infection in humans worldwide. Oxidative stress and gastric inflammation are involved in the progression of HP-related gastric diseases, and they can be targeted by integrating conventional antibiotic treatment with polyphenol-enriched natural products. In this work, we characterised three different propolis extracts and evaluated their stability under in vitro simulated gastric digestion, compared to their main constituents alone. The extract with the highest stability to digestion (namely, the dark propolis extract, DPE) showed a minimum bactericidal concentration (MBC) lower than 1 mg/mL on HP strains with different virulence factors. Finally, since urease is one of the virulence factors contributing to the establishment of a microenvironment that promotes HP infection, we evaluated the possible inhibition of this enzyme by using molecular docking simulations and in vitro colorimetric assay, showing that galangin and pinocembrin may be involved in this activity.
Collapse
Affiliation(s)
- Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulia Romagnoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Paola Albanese
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Federico Rossi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marco Biagi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| |
Collapse
|
27
|
Kavishvar D, Ramachandran A. The yielding behaviour of human mucus. Adv Colloid Interface Sci 2023; 322:103049. [PMID: 38039907 DOI: 10.1016/j.cis.2023.103049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/03/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023]
Abstract
Mucus is a viscoelastic material with non-linear rheological properties such as a yield stress of the order of a few hundreds of millipascals to a few tens of pascals, due to a complex network of mucins in water along with non-mucin proteins, DNA and cell debris. In this review, we discuss the origin of the yield stress in human mucus, the changes in the rheology of mucus with the occurrence of diseases, and possible clinical applications in disease detection as well as cure. We delve into the domain of mucus rheology, examining both macro- and microrheology. Macrorheology involves investigations conducted at larger length scales (∼ a few hundreds of μm or higher) using traditional rheometers, which probe properties on a bulk scale. It is significant in elucidating various mucosal functions within the human body. This includes rejecting unwanted irritants out of lungs through mucociliary and cough clearance, protecting the stomach wall from the acidic environment as well as biological entities, safeguarding cervical canal from infections and providing a swimming medium for sperms. Additionally, we explore microrheology, which encompasses studies performed at length scales ranging from a few tens of nm to a μm. These microscale studies find various applications, including the context of drug delivery. Finally, we employ scaling analysis to elucidate a few examples in lung, cervical, and gastric mucus, including settling of irritants in lung mucus, yielding of lung mucus in cough clearance and cilial beating, spreading of exogenous surfactants over yielding mucus, swimming of Helicobacter pylori through gastric mucus, and lining of protective mucus in the stomach. The scaling analyses employed on the applications mentioned above provide us with a deeper understanding of the link between the rheology and the physiology of mucus.
Collapse
Affiliation(s)
- Durgesh Kavishvar
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| | - Arun Ramachandran
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Chen C, Beloqui A, Xu Y. Oral nanomedicine biointeractions in the gastrointestinal tract in health and disease. Adv Drug Deliv Rev 2023; 203:115117. [PMID: 37898337 DOI: 10.1016/j.addr.2023.115117] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/03/2023] [Accepted: 10/21/2023] [Indexed: 10/30/2023]
Abstract
Oral administration is the preferred route of administration based on the convenience for and compliance of the patient. Oral nanomedicines have been developed to overcome the limitations of free drugs and overcome gastrointestinal (GI) barriers, which are heterogeneous across healthy and diseased populations. This review aims to provide a comprehensive overview and comparison of the oral nanomedicine biointeractions in the gastrointestinal tract (GIT) in health and disease (GI and extra-GI diseases) and highlight emerging strategies that exploit these differences for oral nanomedicine-based treatment. We introduce the key GI barriers related to oral delivery and summarize their pathological changes in various diseases. We discuss nanomedicine biointeractions in the GIT in health by describing the general biointeractions based on the type of oral nanomedicine and advanced biointeractions facilitated by advanced strategies applied in this field. We then discuss nanomedicine biointeractions in different diseases and explore how pathological characteristics have been harnessed to advance the development of oral nanomedicine.
Collapse
Affiliation(s)
- Cheng Chen
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Ana Beloqui
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium; WEL Research Institute, avenue Pasteur, 6, 1300 Wavre, Belgium.
| | - Yining Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
29
|
Su-Arcaro C, Liao W, Bieniek K, Constantino MA, Decker SM, Turner BS, Bansil R. Unraveling the Intertwined Effect of pH on Helicobacter pylori Motility and the Microrheology of the Mucin-Based Medium It Swims in. Microorganisms 2023; 11:2745. [PMID: 38004756 PMCID: PMC10673263 DOI: 10.3390/microorganisms11112745] [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: 08/16/2023] [Revised: 10/16/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
The gastric pathogen, Helicobacter pylori bacteria have to swim across a pH gradient from 2 to 7 in the mucus layer to colonize the gastric epithelium. Previous studies from our group have shown that porcine gastric mucin (PGM) gels at an acidic pH < 4, and H. pylori bacteria are unable to swim in the gel, although their flagella rotate. Changing pH impacts both the rheological properties of gastric mucin and also influences the proton (H+)-pumped flagellar motors of H. pylori as well as their anti-pH sensing receptors. To unravel these intertwined effects of acidic pH on both the viscoelastic properties of the mucin-based mucus as well as the flagellar motors and chemo-receptors of the bacterium, we compared the motility of H. pylori in PGM with that in Brucella broth (BB10) at different pH values using phase contrast microscopy to track the motion of the bacteria. The results show that the distribution of swimming speeds and other characteristics of the bacteria trajectories exhibit pH-dependent differences in both media. The swimming speed exhibits a peak at pH 4 in BB10, and a less pronounced peak at a higher pH of 5 in PGM. At all pH values, the bacteria swam faster and had a longer net displacement in BB10 compared to PGM. While the bacteria were stuck in PGM gels at pH < 4, they swam at these acidic pH values in BB10, although with reduced speed. Decreasing pH leads to a decreased fraction of motile bacteria, with a decreased contribution of the faster swimmers to the distributions of speeds and net displacement of trajectories. The body rotation rate is weakly dependent on pH in BB10, whereas in PGM bacteria that are immobilized in the low pH gel are capable of mechano-sensing and rotate faster. Bacteria can be stuck in the gel in various ways, including the flagella getting entangled in the fibers of the gel or the cell body being stuck to the gel. Our results show that in BB10, swimming is optimized at pH4, reflecting the combined effects of pH sensing by anti-pH tactic receptors and impact on H+ pumping of flagellar motors, while the increase in viscosity of PGM with decreasing pH and gelation below pH 4 lead to further reduction in swimming speed, with optimal swimming at pH 5 and immobilization of bacteria below pH 4.
Collapse
Affiliation(s)
- Clover Su-Arcaro
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| | - Wentian Liao
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| | - Katarzyna Bieniek
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| | - Maira A. Constantino
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| | - Savannah M. Decker
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| | - Bradley S. Turner
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Rama Bansil
- Department of Physics, Boston University, Boston, MA 02215, USA; (C.S.-A.); (W.L.); (K.B.); (M.A.C.); (S.M.D.)
| |
Collapse
|
30
|
Moosavian M, Kushki E, Navidifar T, Hajiani E, Mandegari M. Is There a Real Relationship between the Presence of Helicobacter pylori in Dental Plaque and Gastric Infection? A Genotyping and Restriction Fragment Length Polymorphism Study on Patient Specimens with Dyspepsia in Southwest Iran. Int J Microbiol 2023; 2023:1212009. [PMID: 38021088 PMCID: PMC10645488 DOI: 10.1155/2023/1212009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 07/26/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background The oral cavity can act as an extra gastric reservoir for H. pylori, and the presence of the bacteria in the oral cavity is associated with a higher risk of dental caries development. This study aimed to determine the genotype and evaluate the association between the presence of H. pylori in dental plaque and gastric biopsy specimens in dyspeptic patients in Ahvaz, Southwest Iran. Methods In this study, 106 patients with recruited dyspeptic complaints were selected, and from each patient, two gastric antral biopsy specimens and two dental plagues were examined. The presence of H. pylori was identified by the rapid urease test (RUT) and the amplification of ureAB and 16S rRNA genes. Also, to verify a hypothetical mouth-to-stomach infection route, the enzymatic digestions of three genes of cagA, vacA, and ureAB in H. pylori strains isolated from dental plaques and stomach samples were compared for each same case. Results H. pylori was found in the stomach of 52.8% (56/106) and the dental plaques of 17.9% (19/106) of the studied cases. On the other hand, H. pylori was recognized in the stomach of all 19 cases with oral colonization. Following a combination of restriction fragment lengths 21 polymorphism (RFLP) patterns of these three known genes on stomach and dental plague samples, 14 and 11 unique patterns were seen, respectively. However, for all H. pylori-positive cases (19), the comparison of RLFP patterns of these genes in dental plaque and gastric biopsy specimens was different for the same case. Conclusions In this study, it seems that there is no significant association between the presence of H. pylori in dental plaque and the stomach of the same case.
Collapse
Affiliation(s)
- Mojtaba Moosavian
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elyas Kushki
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tahereh Navidifar
- Department of Microbiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Eskandar Hajiani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahdi Mandegari
- School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
31
|
Donahue R, Sahoo JK, Rudolph S, Chen Y, Kaplan DL. Mucosa-Mimetic Materials for the Study of Intestinal Homeostasis and Disease. Adv Healthc Mater 2023; 12:e2300301. [PMID: 37329337 DOI: 10.1002/adhm.202300301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Mucus is a viscoelastic hydrogel that lines and protects the epithelial surfaces of the body that houses commensal microbiota and functions in host defense against pathogen invasion. As a first-line physical and biochemical barrier, intestinal mucus is involved in immune surveillance and spatial organization of the microbiome, while dysfunction of the gut mucus barrier is implicated in several diseases. Mucus can be collected from a variety of mammalian sources for study, however, established methods are challenging in terms of scale and efficiency, as well as with regard to rheological similarity to native human mucus. Therefore, there is a need for mucus-mimetic hydrogels that more accurately reflect the physical and chemical profile of the in vivo human epithelial environment to enable the investigation of the role of mucus in human disease and interactions with the intestinal microbiome. This review will evaluate the material properties of synthetic mucus mimics to date designed to address the above need, with a focus toward an improved understanding of the biochemical and immunological functions of these biopolymers related to utility for research and therapeutic applications.
Collapse
Affiliation(s)
- Rebecca Donahue
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
32
|
Bustos NA, Ribbeck K, Wagner CE. The role of mucosal barriers in disease progression and transmission. Adv Drug Deliv Rev 2023; 200:115008. [PMID: 37442240 DOI: 10.1016/j.addr.2023.115008] [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: 08/31/2022] [Revised: 05/22/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Mucus is a biological hydrogel that coats and protects all non-keratinized wet epithelial surfaces. Mucins, the primary structural components of mucus, are critical components of the gel layer that protect against invading pathogens. For communicable diseases, pathogen-mucin interactions contribute to the pathogen's fate and the potential for disease progression in-host, as well as the potential for onward transmission. We begin by reviewing in-host mucus filtering mechanisms, including size filtering and interaction filtering, which regulate the permeability of mucus barriers to all molecules including pathogens. Next, we discuss the role of mucins in communicable diseases at the point of transmission (i.e. how the encapsulation of pathogens in emitted mucosal droplets externally to hosts may modulate pathogen infectivity and viability). Overall, mucosal barriers modulate both host susceptibility as well as the dynamics of population-level disease transmission. The study of mucins and their use in models and experimental systems are therefore crucial for understanding the mechanistic biophysical principles underlying disease transmission and the early stages of host infection.
Collapse
Affiliation(s)
- Nicole A Bustos
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline E Wagner
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
33
|
Wang CM, Fernez MT, Woolston BM, Carrier RL. Native gastrointestinal mucus: Critical features and techniques for studying interactions with drugs, drug carriers, and bacteria. Adv Drug Deliv Rev 2023; 200:114966. [PMID: 37329985 PMCID: PMC11184232 DOI: 10.1016/j.addr.2023.114966] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Gastrointestinal mucus plays essential roles in modulating interactions between intestinal lumen contents, including orally delivered drug carriers and the gut microbiome, and underlying epithelial and immune tissues and cells. This review is focused on the properties of and methods for studying native gastrointestinal mucus and its interactions with intestinal lumen contents, including drug delivery systems, drugs, and bacteria. The properties of gastrointestinal mucus important to consider in its analysis are first presented, followed by a discussion of different experimental setups used to study gastrointestinal mucus. Applications of native intestinal mucus are then described, including experimental methods used to study mucus as a barrier to drug delivery and interactions with intestinal lumen contents that impact barrier properties. Given the significance of the microbiota in health and disease, its impact on drug delivery and drug metabolism, and the use of probiotics and microbe-based delivery systems, analysis of interactions of bacteria with native intestinal mucus is then reviewed. Specifically, bacteria adhesion to, motility within, and degradation of mucus is discussed. Literature noted is focused largely on applications of native intestinal mucus models as opposed to isolated mucins or reconstituted mucin gels.
Collapse
Affiliation(s)
- Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
34
|
Lim KPK, Lee AJL, Jiang X, Teng TZJ, Shelat VG. The link between Helicobacter pylori infection and gallbladder and biliary tract diseases: A review. Ann Hepatobiliary Pancreat Surg 2023; 27:241-250. [PMID: 37357161 PMCID: PMC10472116 DOI: 10.14701/ahbps.22-056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 06/27/2023] Open
Abstract
Helicobacter pylori is a gram-negative pathogen commonly associated with peptic ulcer disease and gastric cancer. H. pylori infection has also been reported in cholelithiasis, cholecystitis, gallbladder polyps, and biliary tract cancers. However, the association between H. pylori and gallbladder and biliary tract pathologies remains unclear due to the paucity of literature. In response to the current literature gap, we aim to review and provide an updated summary of the association between H. pylori with gallbladder and biliary tract diseases and its impact on their clinical management. Relevant peer-reviewed studies were retrieved from Medline, PubMed, Embase, and Cochrane databases. We found that H. pylori infection was associated with cholelithiasis, chronic cholecystitis, biliary tract cancer, primary sclerosing cholangitis, and primary biliary cholangitis but not with gallbladder polyps. While causal links have been reported, prospective longitudinal studies are required to conclude the association between H. pylori and gallbladder pathologies. Clinicians should be aware of the implications that H. pylori infection has on the management of these diseases.
Collapse
Affiliation(s)
- Klay Puay Khim Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Aaron Jia Loong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Xiuting Jiang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Thomas Zheng Jie Teng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of General Surgery, Tan Tock Seng Hospital, Singapore
| | - Vishal G. Shelat
- Department of General Surgery, Tan Tock Seng Hospital, Singapore
| |
Collapse
|
35
|
Mu T, Lu ZM, Wang WW, Feng H, Jin Y, Ding Q, Wang LF. Helicobacter pylori intragastric colonization and migration: Endoscopic manifestations and potential mechanisms. World J Gastroenterol 2023; 29:4616-4627. [PMID: 37662858 PMCID: PMC10472897 DOI: 10.3748/wjg.v29.i30.4616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/01/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
After being ingested and entering the human stomach, Helicobacter pylori (H. pylori) adopts several effective strategies to adhere to and colonize the gastric mucosa and move to different regions of the stomach to obtain more nutrients and escape from the harsher environments of the stomach, leading to acute infection and chronic gastritis, which is the basis of malignant gastric tumors. The endoscopic manifestations and pathological features of H. pylori infection are diverse and vary with the duration of infection. In this review, we describe the endoscopic manifestations of each stage of H. pylori gastritis and then reveal the potential mechanisms of bacterial intragastric colonization and migration from the perspective of endoscopists to provide direction for future research on the effective therapy and management of H. pylori infection.
Collapse
Affiliation(s)
- Tong Mu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Zhi-Ming Lu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Wen-Wen Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Hua Feng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Yan Jin
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Qian Ding
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Li-Fen Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| |
Collapse
|
36
|
Sahoo R, Chakrabarti R. Structure and dynamics of an active polymer chain inside a nanochannel grafted with polymers. SOFT MATTER 2023; 19:5978-5988. [PMID: 37497754 DOI: 10.1039/d3sm00618b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
We use computer simulations to investigate the complex dynamics of a polymer, made of active Brownian particles, inside a channel grafted internally with passive polymer chains. Our simulations reveal that this probe-polymer, if passive, exhibits a compact structure when its interaction is repulsive with the grafted chains as it tends to stay within the hollow space created along the axis of the channel. On increasing the attractive interaction, the passive probe-polymer is pulled towards the grafted polymeric region and adopts an extended structure. By contrast, switching on the activity helps the probe-polymer to escape from the local traps caused by the sticky grafted chains. The interplay between the activity of the probe-polymer and its sticky interaction with the grafted chains results in shrinking, followed by swelling as the activity is increased. To elucidate the dynamics we compute the mean square displacement (MSD) of the center of mass of the probe-polymer, which increases monotonically with activity and displays superdiffusive behavior at an intermediate time and enhanced diffusion at a long time period. In addition, compared with the attractive interaction, the active probe-polymer shows faster dynamics when the interaction is repulsive to the grafted polymers. We believe that our current study will provide insights into the structural changes and dynamics of active polymers in heterogeneous media and will be useful in designing polymer-based drug delivery vehicles.
Collapse
Affiliation(s)
- Rajiblochan Sahoo
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| | - Rajarshi Chakrabarti
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| |
Collapse
|
37
|
Medina S, Miller M. Synthetic Colonic Mucus Enables the Development of Modular Microbiome Organoids. RESEARCH SQUARE 2023:rs.3.rs-3164407. [PMID: 37577510 PMCID: PMC10418553 DOI: 10.21203/rs.3.rs-3164407/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The human colon is home to more than a trillion microorganisms that modulate diverse gastrointestinal processes and pathophysiologies. Our understanding of how this gut ecosystem impacts human health, although evolving, is still in its nascent stages and has been slowed by the lack of accessible and scalable tools suitable to studying complex host-mucus-microbe interactions. In this work, we report a synthetic gel-like material capable of recapitulating the varied structural, mechanical, and biochemical profiles of native human colonic mucus to develop compositionally simple microbiome screening platforms with broad utility in microbiology and drug discovery. The viscous fibrillar material is realized through the templated assembly of a fluorine-rich amino acid at liquid-liquid phase separated interfaces. The fluorine-assisted mucus surrogate (FAMS) can be decorated with various mucins to serve as a habitat for microbial colonization and be integrated with human colorectal epithelial cells to generate multicellular artificial mucosae, which we refer to as a microbiome organoid. Notably, FAMS are made with inexpensive and commercially available materials, and can be generated using simple protocols and standard laboratory hardware. As a result, this platform can be broadly incorporated into various laboratory settings to advance our understanding of probiotic biology and inform in vivo approaches. If implemented into high throughput screening approaches, FAMS may represent a valuable tool in drug discovery to study compound metabolism and gut permeability, with an exemplary demonstration of this utility presented here.
Collapse
|
38
|
Woodhams DC, McCartney J, Walke JB, Whetstone R. The adaptive microbiome hypothesis and immune interactions in amphibian mucus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104690. [PMID: 37001710 PMCID: PMC10249470 DOI: 10.1016/j.dci.2023.104690] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 05/20/2023]
Abstract
The microbiome is known to provide benefits to hosts, including extension of immune function. Amphibians are a powerful immunological model for examining mucosal defenses because of an accessible epithelial mucosome throughout their developmental trajectory, their responsiveness to experimental treatments, and direct interactions with emerging infectious pathogens. We review amphibian skin mucus components and describe the adaptive microbiome as a novel process of disease resilience where competitive microbial interactions couple with host immune responses to select for functions beneficial to the host. We demonstrate microbiome diversity, specificity of function, and mechanisms for memory characteristic of an adaptive immune response. At a time when industrialization has been linked to losses in microbiota important for host health, applications of microbial therapies such as probiotics may contribute to immunotherapeutics and to conservation efforts for species currently threatened by emerging diseases.
Collapse
Affiliation(s)
- Douglas C Woodhams
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA.
| | - Julia McCartney
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Jenifer B Walke
- Department of Biology, Eastern Washington University, Cheney, WA, 99004-2440, USA
| | - Ross Whetstone
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| |
Collapse
|
39
|
Huang H, Zhong W, Wang X, Yang Y, Wu T, Chen R, Liu Y, He F, Li J. The role of gastric microecological dysbiosis in gastric carcinogenesis. Front Microbiol 2023; 14:1218395. [PMID: 37583514 PMCID: PMC10423824 DOI: 10.3389/fmicb.2023.1218395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Gastric cancer (GC) is the leading cause of cancer-related death worldwide, and reducing its mortality has become an urgent public health issue. Gastric microecological dysbiosis (including bacteria, fungi, viruses, acid suppressants, antibiotics, and surgery) can lead to gastric immune dysfunction or result in a decrease in dominant bacteria and an increase in the number and virulence of pathogenic microorganisms, which in turn promotes development of GC. This review analyzes the relationship between gastric microecological dysbiosis and GC, elucidates dynamic alterations of the microbiota in Correa's cascade, and identifies certain specific microorganisms as potential biomarkers of GC to aid in early screening and diagnosis. In addition, this paper presents the potential of gastric microbiota transplantation as a therapeutic target for gastric cancer, providing a new direction for future research in this field.
Collapse
Affiliation(s)
- Hui Huang
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Wei Zhong
- Chengdu Medical College, Chengdu, Sichuan, China
| | | | - Ying Yang
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Tianmu Wu
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Runyang Chen
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Yanling Liu
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Feng He
- Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Jun Li
- Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Xu Y, Walduck AK, Pan H. Editorial: The pathogenesis and treatment of Helicobacter pylori-induced diseases. Front Cell Infect Microbiol 2023; 13:1219503. [PMID: 37469603 PMCID: PMC10352097 DOI: 10.3389/fcimb.2023.1219503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Affiliation(s)
- Yifei Xu
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Anna K. Walduck
- Rural Health Research Institute, Charles Sturt University, Orange, NSW, Australia
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
41
|
Gupta N, Kumar A, Verma VK. Strategies adopted by gastric pathogen Helicobacter pylori for a mature biofilm formation: Antimicrobial peptides as a visionary treatment. Microbiol Res 2023; 273:127417. [PMID: 37267815 DOI: 10.1016/j.micres.2023.127417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 06/04/2023]
Abstract
Enormous efforts in recent past two decades to eradicate the pathogen that has been prevalent in half of the world's population have been problematic. The biofilm formed by Helicobacter pylori provides resistance towards innate immune cells, various combinatorial antibiotics, and human antimicrobial peptides, despite the fact that these all are potent enough to eradicate it in vitro. Biofilm provides the opportunity to secrete various virulence factors that strengthen the interaction between host and pathogen helping in evading the innate immune system and ultimately leading to persistence. To our knowledge, this review is the first of its kind to explain briefly the journey of H. pylori starting with the chemotaxis, the mechanism for selecting the site for colonization, the stress faced by the pathogen, and various adaptations to evade these stress conditions by forming biofilm and the morphological changes acquired by the pathogen in mature biofilm. Furthermore, we have explained the human GI tract antimicrobial peptides and the reason behind the failure of these AMPs, and how encapsulation of Pexiganan-A(MSI-78A) in a chitosan microsphere increases the efficiency of eradication.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Marg, New Delhi 110021, India.
| | - Atul Kumar
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Marg, New Delhi 110021, India
| | - Vijay Kumar Verma
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Marg, New Delhi 110021, India.
| |
Collapse
|
42
|
Bugaytsova JA, Moonens K, Piddubnyi A, Schmidt A, Edlund JO, Lisiutin G, Brännström K, Chernov YA, Thorel K, Tkachenko I, Sharova O, Vikhrova I, Butsyk A, Shubin P, Chyzhma R, Johansson DX, Marcotte H, Sjöström R, Shevtsova A, Bylund G, Rakhimova L, Lundquist A, Berhilevych O, Kasianchuk V, Loboda A, Ivanytsia V, Hultenby K, Persson MAA, Gomes J, Matos R, Gartner F, Reis CA, Whitmire JM, Merrell DS, Pan-Hammarström Q, Landström M, Oscarson S, D’Elios MM, Agreus L, Ronkainen J, Aro P, Engstrand L, Graham DY, Kachkovska V, Mukhopadhyay A, Chaudhuri S, Karmakar BC, Paul S, Kravets O, Camorlinga M, Torres J, Berg DE, Moskalenko R, Haas R, Remaut H, Hammarström L, Borén T. Helicobacter pylori attachment-blocking antibodies protect against duodenal ulcer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542096. [PMID: 37292721 PMCID: PMC10245814 DOI: 10.1101/2023.05.24.542096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.
Collapse
Affiliation(s)
- Jeanna A. Bugaytsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
| | - Kristof Moonens
- Structural and Molecular Microbiology, VIB Department of Structural Biology, VIB, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Present address: Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Artem Piddubnyi
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Alexej Schmidt
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
- Present address: Department of Medical Biosciences, Umeå University, SE90185 Umeå, Sweden
| | - Johan Olofsson Edlund
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- The Biochemical Imaging Center Umeå (BICU), Umeå University, SE90187 Umeå, Sweden
| | - Gennadii Lisiutin
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Microbiology, Virology and Biotechnology, Odesa Mechnikov National University, 65082 Odesa, Ukraine
| | - Kristoffer Brännström
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- The Biochemical Imaging Center Umeå (BICU), Umeå University, SE90187 Umeå, Sweden
- Present address: Pfizer Worldwide R&D, BioMedicine Design, 10 555 Science Center Drive, San Diego CA, 92121 USA
| | - Yevgen A. Chernov
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Kaisa Thorel
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Iryna Tkachenko
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Oleksandra Sharova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Iryna Vikhrova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Anna Butsyk
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Pavlo Shubin
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Ruslana Chyzhma
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Daniel X. Johansson
- Department of Clinical Neuroscience, Karolinska Institutet at Center for Molecular Medicine, Karolinska University Hospital, Solna, SE17176 Stockholm, Sweden
| | - Harold Marcotte
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Rolf Sjöström
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Anna Shevtsova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Göran Bylund
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
| | - Lena Rakhimova
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- Present address: Department of Odontology, Umeå University, SE90187 Umeå, Sweden
| | - Anders Lundquist
- Department of Statistics, USBE, Umeå University, SE90187 Umeå, Sweden
- Umeå Center for Functional Brain Imaging, Umeå University, SE90187 Umeå, Sweden
| | - Oleksandra Berhilevych
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Victoria Kasianchuk
- Department of Public Health, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Andrii Loboda
- Department of Pediatrics, Medical Institute, Sumy State University, 40018 Sumy, Ukraine
| | - Volodymyr Ivanytsia
- Department of Microbiology, Virology and Biotechnology, Odesa Mechnikov National University, 65082 Odesa, Ukraine
| | - Kjell Hultenby
- Departments of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet at Karolinska University Hospital, SE14186 Huddinge, Sweden
| | - Mats A. A. Persson
- Department of Clinical Neuroscience, Karolinska Institutet at Center for Molecular Medicine, Karolinska University Hospital, Solna, SE17176 Stockholm, Sweden
| | - Joana Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Rita Matos
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Fátima Gartner
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Celso A. Reis
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | | | - D. Scott Merrell
- Department of Microbiology and Immunology, USUHS, Bethesda, MD 20814, USA
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Maréne Landström
- Present address: Department of Medical Biosciences, Umeå University, SE90185 Umeå, Sweden
| | - Stefan Oscarson
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Mario M. D’Elios
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134 Firenze, Italy
| | - Lars Agreus
- Division of Family Medicine and Primary Care, Karolinska Institutet, SE14183 Huddinge, Sweden
| | - Jukka Ronkainen
- University of Oulu, Center for Life Course Health Research and Primary Health Care Center, Tornio Finland
| | - Pertti Aro
- University of Oulu, Center for Life Course Health Research and Primary Health Care Center, Tornio Finland
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE17177 Stockholm, Sweden
- Present address: Science for Life Laboratory, SE17165, Solna, Sweden
| | - David Y. Graham
- Department of Medicine, Molecular Virology and Microbiology, Baylor College of Medicine, Michael E. DeBakey VAMC, 2002 Holcombe Blvd. Houston, TX, 77030 USA
| | - Vladyslava Kachkovska
- Department of Internal Medicine, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Asish Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Sujit Chaudhuri
- Department of Gastroenterology, AMRI Hospital, Salt Lake City. Kolkata, West Bengal 700098, India
| | - Bipul Chandra Karmakar
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Sangita Paul
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases P 33, CIT Road, Scheme XM, Kolkata 700010, India
| | - Oleksandr Kravets
- Department of Surgery, Traumatology, Orthopedics and Physiology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Margarita Camorlinga
- Unidad de Investigacion en Enfermedades Infecciosas, UMAE Pediatria, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Javier Torres
- Unidad de Investigacion en Enfermedades Infecciosas, UMAE Pediatria, CMN SXXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Douglas E. Berg
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Roman Moskalenko
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Department of Pathology, Medical Institute, Sumy State University, 40007 Sumy, Ukraine
| | - Rainer Haas
- German Center for Infection Research (DZIF), Munich Site, 80336 Munich, Germany
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer-Institute, Faculty of Medicine, LMU Munich, Germany
| | - Han Remaut
- Structural and Molecular Microbiology, VIB Department of Structural Biology, VIB, 1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Lennart Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, SE14183, Huddinge, Sweden
| | - Thomas Borén
- Department of Medical Biochemistry and Biophysics, Umeå University, SE90187 Umeå, Sweden
- SUMEYA, The Ukrainian-Swedish Research Center, Sumy State University, 40022 Sumy, Ukraine
- Lead contact
| |
Collapse
|
43
|
Yang H, Wang L, Zhang M, Hu B. The Role of Adhesion in Helicobacter pylori Persistent Colonization. Curr Microbiol 2023; 80:185. [PMID: 37071212 DOI: 10.1007/s00284-023-03264-6] [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: 11/12/2022] [Accepted: 03/10/2023] [Indexed: 04/19/2023]
Abstract
Helicobacter pylori (H. pylori) has coevolved with its human host for more than 100 000 years. It can safely colonize around the epithelium of gastric glands via their specific microstructures and proteins. Unless patients receive eradication treatment, H. pylori infection is always lifelong. However, few studies have discussed the reasons. This review will focus on the adhesion of H. pylori from the oral cavity to gastric mucosa and summarize the possible binding and translocation characteristics. Adhesion is the first step for persistent colonization after the directional motility, and factors related to adhesion are necessary. Outer membrane proteins, such as the blood group antigen binding adhesin (BabA) and the sialic acid binding adhesin (SabA), play pivotal roles in binding to human mucins and cellular surfaces. And this may offer different perspectives on eradication.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Wu Hou District, Chengdu City, 610041, Sichuan Province, China
| | - Lixia Wang
- The Second Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Miao Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Wu Hou District, Chengdu City, 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Wu Hou District, Chengdu City, 610041, Sichuan Province, China.
| |
Collapse
|
44
|
Nazari F, Shoele K, Mohammadigoushki H. Helical Locomotion in Yield Stress Fluids. PHYSICAL REVIEW LETTERS 2023; 130:114002. [PMID: 37001094 DOI: 10.1103/physrevlett.130.114002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 01/27/2023] [Indexed: 06/19/2023]
Abstract
We report three stages for locomotion of a helical swimmer in yield stress fluids. In the first stage, the swimmer must overcome the material's yield strain to generate rotational motion. However, exceeding the first threshold is not sufficient for locomotion. Only when the viscous forces are sufficiently strong to plastically deform the material to a finite distance away from the swimmer will net locomotion occur. Once locomotion is underway in the third stage, the yield stress retards swimming at small pitch angles. Conversely, at large pitch angles, yield stress dominates the flow by enhancing swimming speed. Flow visualizations reveal a highly localized flow near the swimmer in yield stress fluids.
Collapse
Affiliation(s)
- Farshad Nazari
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, USA
| | - Kourosh Shoele
- Department of Mechanical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, USA
| | - Hadi Mohammadigoushki
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, USA
| |
Collapse
|
45
|
Sheikhshoaei A, Rajabi M. Utilizing passive elements to break time reversibility at low Reynolds number: a swimmer with one activated element. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:15. [PMID: 36929245 DOI: 10.1140/epje/s10189-023-00273-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 02/27/2023] [Indexed: 06/18/2023]
Abstract
In the realm of low Reynolds number, the shape-changing biological and artificial matters need to break time reversibility in the course of their strokes to achieve motility. This necessity is well described in the so-called scallop theorem. In this work, considering low Reynolds number, a novel and versatile swimmer is proposed as an example of a new scheme to break time reversibility kinematically and, in turn, produce net motion. The swimmer consists of one sphere as a cargo or carried body, joined by one activated link with time-varying length, to another perpendicular rigid link, as the support of two passively flapping disks, at its end. The disks are free to rotate between their fixed minimum and maximum angles. The system's motion in two dimensions is simulated, and the maneuverability of the swimmer is discussed. The minimal operating parameters for steering of the swimmer are studied, and the limits of the swimmer are identified. The introduced swimming mechanism can be employed as a simple model system for biological living matters as well as artificial microswimmers.
Collapse
Affiliation(s)
- Amir Sheikhshoaei
- School of Mechanical Engineering, Iran University of Science and Technology, Narmak, Tehran, Iran.
| | - Majid Rajabi
- School of Mechanical Engineering, Iran University of Science and Technology, Narmak, Tehran, Iran
| |
Collapse
|
46
|
Xi J, Li Y, Zhang H, Bai Z. Dynamic variations of the gastric microbiota: Key therapeutic points in the reversal of Correa's cascade. Int J Cancer 2023; 152:1069-1084. [PMID: 36029278 DOI: 10.1002/ijc.34264] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
Correa's cascade is a dynamic process in the development of intestinal-type gastric cancer (GC), and its pathological features, gastric microbiota and interactions between microorganisms and their hosts vary at different developmental stages. The characteristics of cells, tissues and gastric microbiota before or after key therapeutic points are critical for monitoring malignant transformation and early tumour reversal. This review summarises the pathological features of gastric mucosa, characteristics of gastric microbiota, specific microbial markers, microbe-microbe interactions and microbe-host interactions at different stages in Correa's cascade. The markers related to each Correa's cascade point were analysed in detail. We attempted to identify key therapeutic points for early cancer reversal and provide a novel approach to reduce the incidence of GC and improve precise treatment.
Collapse
Affiliation(s)
- Jiahui Xi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, Lanzhou, China
| | - Yonghong Li
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumour, Gansu Provincial Hospital, Lanzhou, China
| | - Hui Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhongtian Bai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, Lanzhou, China.,General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
47
|
d'Amone L, Sahoo JK, Ostrovsky-Snider N, Kaplan DL, Omenetto FG. Boronic Acid-Tethered Silk Fibroin for pH-Dependent Mucoadhesion. Biomacromolecules 2023; 24:1310-1317. [PMID: 36763594 DOI: 10.1021/acs.biomac.2c01349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Mucus lines all surfaces of the human body not covered by skin and provides lubrication, hydration, and protection. The properties of mucus are influenced by changes in pH that may occur due to physiological conditions and pathological circumstances. Reinforcing the mucus barrier with biopolymers that can adhere to mucus in different conditions can be a useful strategy for protecting the underlying mucosae from damage. In this work, regenerated silk fibroin (silk) was chemically modified with phenyl boronic acid to form reversible covalent complexes with the 1,2- or 1,3-diols. The silk modified with boronic acid pendant groups has an increased affinity for mucins, whose carbohydrate component is rich in diols. These results offer new applications of silk in mucoadhesion, and the ability to bind diols to the silk lays the foundation for the development of silk-based sugar-sensing platforms.
Collapse
Affiliation(s)
- Luciana d'Amone
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
| | - Fiorenzo G Omenetto
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02153, United States
- Department of Electrical and Computer Engineering, Tufts University, Medford, Massachusetts 02153, United States
- Department of Physics, Tufts University, Medford, Massachusetts 02153, United States
- Laboratory for Living Devices, Tufts University, Medford, Massachusetts 02153, United States
| |
Collapse
|
48
|
Bansil R, Constantino MA, Su-Arcaro C, Liao W, Shen Z, Fox JG. Motility of Different Gastric Helicobacter spp. Microorganisms 2023; 11:634. [PMID: 36985208 PMCID: PMC10058440 DOI: 10.3390/microorganisms11030634] [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: 02/07/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Helicobacter spp., including the well-known human gastric pathogen H. pylori, can cause gastric diseases in humans and other mammals. They are Gram-negative bacteria that colonize the gastric epithelium and use their multiple flagella to move across the protective gastric mucus layer. The flagella of different Helicobacter spp. vary in their location and number. This review focuses on the swimming characteristics of different species with different flagellar architectures and cell shapes. All Helicobacter spp. use a run-reverse-reorient mechanism to swim in aqueous solutions, as well as in gastric mucin. Comparisons of different strains and mutants of H. pylori varying in cell shape and the number of flagella show that their swimming speed increases with an increasing number of flagella and is somewhat enhanced with a helical cell body shape. The swimming mechanism of H. suis, which has bipolar flagella, is more complex than that of unipolar H. pylori. H. suis exhibits multiple modes of flagellar orientation while swimming. The pH-dependent viscosity and gelation of gastric mucin significantly impact the motility of Helicobacter spp. In the absence of urea, these bacteria do not swim in mucin gel at pH < 4, even though their flagellar bundle rotates.
Collapse
Affiliation(s)
- Rama Bansil
- Department of Physics, Boston University, Boston, MA 02215, USA
| | | | | | - Wentian Liao
- Department of Physics, Boston University, Boston, MA 02215, USA
| | - Zeli Shen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02138, USA
| | - James G. Fox
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02138, USA
| |
Collapse
|
49
|
Freire Haddad H, Roe EF, Collier JH. Expanding opportunities to engineer mucosal vaccination with biomaterials. Biomater Sci 2023; 11:1625-1647. [PMID: 36723064 DOI: 10.1039/d2bm01694j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal vaccines are receiving increasing interest both for protecting against infectious diseases and for inducing therapeutic immune responses to treat non-infectious diseases. However, the mucosal barriers of the lungs, gastrointestinal tract, genitourinary tract, nasal, and oral tissues each present unique challenges for constructing efficacious vaccines. Vaccination through each of these mucosae requires transport through the mucus and across specialized epithelia to reach tissue-specific immune cells and lymphoid structures, necessitating finely tuned and multifunctional strategies. Serving as inspiration for mucosal vaccine design, pathogens have evolved elaborate, diverse, and multipronged approaches to penetrate and infect mucosae. This review is focused on biomaterials-based strategies, many inspired by pathogens, for designing mucosal vaccine platforms. Passive and active technologies are discussed, along with the microbial processes that they seek to mimic.
Collapse
Affiliation(s)
- Helena Freire Haddad
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Emily F Roe
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Joel H Collier
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
50
|
Nikolić I, Chua EG, Tay ACY, Kostrešević A, Pavlović B, Jončić Savić K. Savory, Oregano and Thyme Essential Oil Mixture (HerbELICO ®) Counteracts Helicobacter pylori. Molecules 2023; 28:molecules28052138. [PMID: 36903396 PMCID: PMC10003975 DOI: 10.3390/molecules28052138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Fifty percent of the world's population is infected with Helicobacter pylori, which can trigger many gastrointestinal disorders. H. pylori eradication therapy consists of two to three antimicrobial medicinal products, but they exhibit limited efficacy and may cause adverse side effects. Alternative therapies are urgent. It was assumed that an essential oil mixture, obtained from species from genera Satureja L., Origanum L. and Thymus L. and called the HerbELICO® essential oil mixture, could be useful in H. pylori infection treatment. HerbELICO® was analyzed by GC-MS and assessed in vitro against twenty H. pylori clinical strains isolated from patients of different geographical origins and with different antimicrobial medicinal products resistance profiles, and for its ability to penetrate the artificial mucin barrier. A customer case study included 15 users of HerbELICO®liquid/HerbELICO®solid dietary supplements (capsulated HerbELICO® mixture in liquid/solid form). Carvacrol and thymol were the most dominant compounds (47.44% and 11.62%, respectively), together with p-cymene (13.35%) and γ-terpinene (18.20%). The minimum concentration required to inhibit in vitro H. pylori growth by HerbELICO® was 4-5% (v/v); 10 min exposure to HerbELICO® was enough to kill off the examined H. pylori strains, while HerbELICO® was able to penetrate through mucin. A high eradication rate (up to 90%) and acceptance by consumers was observed.
Collapse
Affiliation(s)
- Ivan Nikolić
- Oncology Institute of Vojvodina, Faculty of Medicine Novi Sad, University of Novi Sad, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Eng Guan Chua
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Diseases Research and Training, University of Western Australia, Perth, WA 6009, Australia
| | - Alfred Chin Yen Tay
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Diseases Research and Training, University of Western Australia, Perth, WA 6009, Australia
| | | | - Bojan Pavlović
- Phytonet Ltd., Science Technology Park, Veljka Dugoševića 54, 11000 Belgrade, Serbia
| | - Katarina Jončić Savić
- Phytonet Ltd., Science Technology Park, Veljka Dugoševića 54, 11000 Belgrade, Serbia
- Correspondence: ; Tel.: +381-11-4085118
| |
Collapse
|