1
|
Budzinski L, Kang GU, Riedel R, Sempert T, Lietz L, Maier R, Büttner J, Bochow B, Tordai MT, Shah A, Abbas A, Momtaz T, Krause JL, Kempkens R, Lehman K, Heinz GA, Benken AE, Bartsch S, Necke K, Hoffmann U, Mashreghi MF, Biesen R, Kallinich T, Alexander T, Jessen B, Weidinger C, Siegmund B, Radbruch A, Schirbel A, Moser B, Chang HD. Single-cell microbiota phenotyping reveals distinct disease and therapy-associated signatures in Crohn's disease. Gut Microbes 2025; 17:2452250. [PMID: 39815413 PMCID: PMC11740678 DOI: 10.1080/19490976.2025.2452250] [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: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
IgA-coated fractions of the intestinal microbiota of Crohn's disease (CD) patients have been shown to contain taxa that hallmark the compositional dysbiosis in CD microbiomes. However, the correlation between other cellular properties of intestinal bacteria and disease has not been explored further, especially for features that are not directly driven by the host immune-system, e.g. the expression of surface sugars by bacteria. By sorting and sequencing IgA-coated and lectin-stained fractions from CD patients microbiota and healthy controls, we found that lectin-stained bacteria were distinct from IgA-coated bacteria, but still displayed specific differences between CD and healthy controls. To exploit the discriminatory potential of both, immunoglobulin coated bacteria and the altered surface sugar expression of bacteria in CD, we developed a multiplexed single cell-based analysis approach for intestinal microbiota. By multi-parameter microbiota flow cytometry (mMFC) we characterized the intestinal microbiota of 55 CD patients and 44 healthy controls for 11-parameters in total, comprising host-immunoglobulin coating and the presence of distinct surface sugar moieties. The data were analyzed by machine-learning to assess disease-specific marker patterns in the microbiota phenotype. mMFC captured detailed characteristics of CD microbiota and identified patterns to classify CD patients. In addition, we identified phenotypic signatures in the CD microbiota which not only reflected remission after 6 weeks of anti-TNF treatment, but were also able to predict remission before the start of an adalimumab treatment course in a pilot study. We here present the proof-of-concept demonstrating that multi-parameter single-cell bacterial phenotyping by mMFC could be a novel tool with high translational potential to expand current microbiome investigations by phenotyping of bacteria to identify disease- and therapy-associated cellular alterations and to reveal novel target properties of bacteria for functional assays and therapeutic approaches.
Collapse
Affiliation(s)
- Lisa Budzinski
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Gi-Ung Kang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - René Riedel
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Bioinformatics and Computational Biology, Department of Cardiology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Toni Sempert
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Leonie Lietz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - René Maier
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Janine Büttner
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Bochow
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcell T. Tordai
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aayushi Shah
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Amro Abbas
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Tanisha Momtaz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- School of Pharmacy, BRAC University, Dhaka, Bangladesh
| | - Jannike L. Krause
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robin Kempkens
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Katrin Lehman
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Gitta A. Heinz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anne E. Benken
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefanie Bartsch
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kathleen Necke
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Hoffmann
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Mir-Farzin Mashreghi
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tilmann Kallinich
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Alexander
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bosse Jessen
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program
| | - Andreas Radbruch
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anja Schirbel
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Benjamin Moser
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DRK Kliniken Berlin, Clinic for internal medicine – Gastroenterology, Haematology and Oncology, Nephrology, Centre for chronic gastrointestinal inflammations, Berlin, Germany
| | - Hyun-Dong Chang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
2
|
Yilmaz B, Macpherson AJ. Delving the depths of 'terra incognita' in the human intestine - the small intestinal microbiota. Nat Rev Gastroenterol Hepatol 2025; 22:71-81. [PMID: 39443711 DOI: 10.1038/s41575-024-01000-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The small intestinal microbiota has a crucial role in gastrointestinal health, affecting digestion, immune function, bile acid homeostasis and nutrient metabolism. The challenges of accessibility at this site mean that our knowledge of the small intestinal microbiota is less developed than of the colonic or faecal microbiota. Here, we summarize the features and fluctuations of the microbiota along the small intestinal tract, focusing on humans, and discuss physicochemical factors and assessment methods, including the technical challenges of investigating the low microbial biomass of the proximal small bowel. We highlight the essential protective mechanisms of the small intestine, including motility, the paracellular barrier and mucus, and secretory immunity, to show their roles in limiting excessive exposure of host tissues to microbial metabolites. We address current knowledge gaps, particularly the variability among individuals, the effects of dysbiosis of the small intestinal microbiota on health and how different taxa in small intestinal microbiota could compensate for each other functionally.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| | - Andrew J Macpherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| |
Collapse
|
3
|
Inui T, Kawamura N, Yamamura M, Kubo K, Yamakage H, Satoh-Asahara N, Ogawa Y, Katsuura G. Oral intake of degalactosylated whey protein increases peripheral blood telomere length in young and aged mice. Sci Rep 2024; 14:30859. [PMID: 39730524 DOI: 10.1038/s41598-024-81597-3] [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: 06/12/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024] Open
Abstract
In order to elucidate novel actions of degalactosylated whey protein (D-WP) in comparison with intact whey protein (WP), the effects of oral intake of D-WP on peripheral blood telomere length and telomerase were examined in young and aged mice. In young mice, peripheral blood telomere length was significantly elongated following oral intake of D-WP for 4 weeks. mRNA expression of both telomerase reverse transcriptase (TERT) and telomerase RNA component (TERC) was significantly increased in the peripheral blood following oral intake of D-WP for 4 weeks. In aged mice, peripheral blood telomere length was significantly decreased as compared with that of young mice, and significantly restored to the level of young mice drinking water by the oral intake of D-WP for 4 weeks. The mRNA expression of peripheral blood TERT and TERC mRNA in aged mice significantly decreased as compared with the level in young mice drinking water, and was significantly restored to the level of expression of young mice drinking water by oral intake of D-WP for 4 weeks. These results suggest that D-WP, but not WP, potently increases peripheral blood telomere length accompanied by increased mRNA expression of TERT and TERC in both young and aged mice.
Collapse
Affiliation(s)
- Toshio Inui
- Saisei Mirai Cell Processing Center, Moriguchi, Japan.
- Cancer Immunotherapy Clinic, 6-14-17 Kinda-cho, Moriguchi-shi, Osaka, 570-0011, Japan.
- Kobe Saisei Mirai Clinic, Kobe, Japan.
- Inui Immunotherapy Clinic, Moriguchi, Japan.
- Saisei Pharma, Moriguchi, Japan.
| | - Namiko Kawamura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Kentaro Kubo
- Cancer Immunotherapy Clinic, 6-14-17 Kinda-cho, Moriguchi-shi, Osaka, 570-0011, Japan
| | - Hajime Yamakage
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Goro Katsuura
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, 612-8555, Japan.
| |
Collapse
|
4
|
Sminia TJ, Aalvink S, de Jong H, Tempelaars MH, Zuilhof H, Abee T, de Vos WM, Tytgat HLP, Wennekes T. Probing Peptidoglycan Synthesis in the Gut Commensal Akkermansia Muciniphila with Bioorthogonal Chemical Reporters. Chembiochem 2024; 25:e202400037. [PMID: 38688858 DOI: 10.1002/cbic.202400037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Our gut microbiota directly influences human physiology in health and disease. The myriad of surface glycoconjugates in both the bacterial cell envelope and our gut cells dominate the microbiota-host interface and play a critical role in host response and microbiota homeostasis. Among these, peptidoglycan is the basic glycan polymer offering the cell rigidity and a basis on which many other glycoconjugates are anchored. To directly study peptidoglycan in gut commensals and obtain the molecular insight required to understand their functional activities we need effective techniques like chemical probes to label peptidoglycan in live bacteria. Here we report a chemically guided approach to study peptidoglycan in a key mucin-degrading gut microbiota member of the Verrucomicrobia phylum, Akkermansia muciniphila. Two novel non-toxic tetrazine click-compatible peptidoglycan probes with either a cyclopropene or isonitrile handle allowed for the detection and imaging of peptidoglycan synthesis in this intestinal species.
Collapse
Affiliation(s)
- Tjerk J Sminia
- Laboratory of Organic Chemistry, Wageningen University and Research, Wageningen, The, Netherlands
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The, Netherlands
| | - Hanna de Jong
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The, Netherlands
| | - Marcel H Tempelaars
- Laboratory of Food Microbiology, Wageningen University and Research, Wageningen, The, Netherlands
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University and Research, Wageningen, The, Netherlands
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Tjakko Abee
- Laboratory of Food Microbiology, Wageningen University and Research, Wageningen, The, Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The, Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanne L P Tytgat
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The, Netherlands
- Current address: Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Tom Wennekes
- Laboratory of Organic Chemistry, Wageningen University and Research, Wageningen, The, Netherlands
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The, Netherlands
| |
Collapse
|
5
|
Chettri D, Chirania M, Boro D, Verma AK. Glycoconjugates: Advances in modern medicines and human health. Life Sci 2024; 348:122689. [PMID: 38710281 DOI: 10.1016/j.lfs.2024.122689] [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: 02/02/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Glycans and their glycoconjugates are complex biomolecules that are crucial for various biological processes. Glycoconjugates are found in all domains of life. They are covalently linked to key biomolecules such as proteins and lipids to play a pivotal role in cell signaling, adhesion, and recognition. The diversity of glycan structures and the associated complexity of glycoconjugates is the reason for their role in intricate biosynthetic pathways. Glycoconjugates play an important role in various diseases where they are actively involved in the immune response as well as in the pathogenicity of infectious diseases. In addition, various autoimmune diseases have been linked to glycosylation defects of different biomolecules, making them an important molecule in the field of medicine. The glycoconjugates have been explored for the development of therapeutics and vaccines, representing a breakthrough in medical science. They also hold significance in research studies to understand the mechanisms behind various biological processes. Finally, glycoconjugates have found an emerging role in various industrial and environmental applications which have been discussed here.
Collapse
Affiliation(s)
- Dixita Chettri
- Department of Microbiology, Sikkim University, Gangtok, Sikkim 737102, India
| | - Manisha Chirania
- Department of Microbiology, Sikkim University, Gangtok, Sikkim 737102, India
| | - Deepjyoti Boro
- Department of Microbiology, Sikkim University, Gangtok, Sikkim 737102, India
| | - Anil Kumar Verma
- Department of Microbiology, Sikkim University, Gangtok, Sikkim 737102, India.
| |
Collapse
|
6
|
Kaushal RS, Naik N, Prajapati M, Rane S, Raulji H, Afu NF, Upadhyay TK, Saeed M. Leishmania species: A narrative review on surface proteins with structural aspects involved in host-pathogen interaction. Chem Biol Drug Des 2023; 102:332-356. [PMID: 36872849 DOI: 10.1111/cbdd.14227] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
In tropical and subtropical regions of the world, leishmaniasis is endemic and causes a range of clinical symptoms in people, from severe tegumentary forms (such as cutaneous, mucocutaneous, and diffuse leishmaniasis) to lethal visceral forms. The protozoan parasite of the genus Leishmania causes leishmaniasis, which is still a significant public health issue, according to the World Health Organization 2022. The public's worry about the neglected tropical disease is growing as new foci of the illness arise, which are exacerbated by alterations in behavior, changes in the environment, and an enlarged range of sand fly vectors. Leishmania research has advanced significantly during the past three decades in a few different avenues. Despite several studies on Leishmania, many issues, such as illness control, parasite resistance, parasite clearance, etc., remain unresolved. The key virulence variables that play a role in the pathogenicity-host-pathogen relationship of the parasite are comprehensively discussed in this paper. The important Leishmania virulence factors, such as Kinetoplastid Membrane Protein-11 (KMP-11), Leishmanolysin (GP63), Proteophosphoglycan (PPG), Lipophosphoglycan (LPG), Glycosylinositol Phospholipids (GIPL), and others, have an impact on the pathophysiology of the disease and enable the parasite to spread the infection. Leishmania infection may arise from virulence factors; they are treatable with medications or vaccinations more promptly and might greatly shorten the duration of treatment. Additionally, our research sought to present a modeled structure of a few putative virulence factors that might aid in the development of new chemotherapeutic approaches for the treatment of leishmaniasis. The predicted virulence protein's structure is utilized to design novel drugs, therapeutic targets, and immunizations for considerable advantage from a higher understanding of the host immune response.
Collapse
Affiliation(s)
- Radhey Shyam Kaushal
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, 391760, Gujarat, India
| | - Nidhi Naik
- Department of Microbiology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Maitri Prajapati
- Department of Microbiology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Shruti Rane
- Department of Microbiology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Himali Raulji
- Department of Microbiology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Ngo Festus Afu
- Department of Biochemistry, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, 391760, Gujarat, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Ha'il, P.O. Box 2440, Hail, 81411, Saudi Arabia
| |
Collapse
|
7
|
Rajeswari M, Pola S, Sravani DSL. Nutritional Modulation of Gut Microbiota Alleviates Metabolic and Neurological Disorders. HUMAN MICROBIOME IN HEALTH, DISEASE, AND THERAPY 2023:97-125. [DOI: 10.1007/978-981-99-5114-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Li J, Qu Z, Liu F, Jing H, Pan Y, Guo S, Ho CL. Diet‐Based Microbiome Modulation: You are What You Eat. PRINCIPLES IN MICROBIOME ENGINEERING 2022:1-46. [DOI: 10.1002/9783527825462.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Sanozky-Dawes R, Barrangou R. Lactobacillus, glycans and drivers of health in the vaginal microbiome. MICROBIOME RESEARCH REPORTS 2022; 1:18. [PMID: 38046360 PMCID: PMC10688826 DOI: 10.20517/mrr.2022.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/08/2022] [Accepted: 04/29/2022] [Indexed: 12/05/2023]
Abstract
A microbiome consists of microbes and their genomes, encompassing bacteria, viruses, fungi, protozoa, archaea, and eukaryotes. These elements interact dynamically in the specific environment in which they reside and evolve. In the past decade, studies of various microbiomes have been prevalent in the scientific literature, accounting for the shift from culture-dependent to culture-independent identification of microbes using new high-throughput sequencing technologies that decipher their composition and sometimes provide insights into their functions. Despite tremendous advances in understanding the gut microbiome, relatively little attention has been devoted to the vaginal environment, notably regarding the ubiquity and diversity of glycans which denote the significant role they play in the maintenance of homeostasis. Hopefully, emerging technologies will aid in the determination of what is a healthy vaginal microbiome, and provide insights into the roles of Lactobacillus, glycans and microbiome-related drivers of health and disease.
Collapse
Affiliation(s)
| | - Rodolphe Barrangou
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
10
|
Inui T, Kawamura N, Nakama R, Inui A, Katsuura G. Degalactosylated Whey Protein Suppresses Inflammatory Responses Induced by Lipopolysaccharide in Mice. Front Nutr 2022; 9:852355. [PMID: 35571950 PMCID: PMC9101058 DOI: 10.3389/fnut.2022.852355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
The effects of degalactosylated whey protein on lipopolysaccharide (LPS)-induced inflammatory responses in mice were observed in comparison with intact whey protein. Intraperitoneal administration of both intact and degalactosylated whey proteins for 5 days did not affect body weight and food intake in mice. On day 6, intraperitoneal administration of LPS induced a marked decrease in body weight 4 h later. The LPS-induced decrease in body weight was significantly suppressed by the administration of degalactosylated whey protein, but not intact whey protein. Administration of LPS also significantly increase plasma tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels, which were significantly suppressed by the administration of degalactosylated whey protein, but not intact whey protein. Moreover, the application of degalactosylated whey protein to RAW264.7 cells significantly reduced mRNA expression of toll-like receptor 4 (TLR4) and significantly increased mRNA expression of mitogen-activated protein kinase phosphatase-1 (MKP-1). The marked increased expression of TNF-α and IL-1β in response to LPS in RAW264.7 cells was significantly suppressed by the application of degalactosylated whey protein. These results suggest that degalactosylated whey protein suppresses the effects of LPS in part by decreasing in TLR4 and increasing in MKP-1.
Collapse
Affiliation(s)
- Toshio Inui
- Saisei Mirai Cell Processing Center, Osaka, Japan
- Kobe Saisei Mirai Clinic, Kobe, Japan
- Inui Immunotherapy Clinic, Osaka, Japan
- *Correspondence: Toshio Inui,
| | - Namiko Kawamura
- Drug Discovery of Next-Generation GcMAF, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Riho Nakama
- Drug Discovery of Next-Generation GcMAF, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akio Inui
- Pharmacological Department of Herbal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Goro Katsuura
- Drug Discovery of Next-Generation GcMAF, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
11
|
de Jong H, Wösten MMSM, Wennekes T. Sweet impersonators: Molecular mimicry of host glycans by bacteria. Glycobiology 2022; 32:11-22. [PMID: 34939094 PMCID: PMC8881735 DOI: 10.1093/glycob/cwab104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/03/2021] [Accepted: 09/28/2021] [Indexed: 12/02/2022] Open
Abstract
All bacteria display surface-exposed glycans that can play an important role in their interaction with the host and in select cases mimic the glycans found on host cells, an event called molecular or glycan mimicry. In this review, we highlight the key bacteria that display human glycan mimicry and provide an overview of the involved glycan structures. We also discuss the general trends and outstanding questions associated with human glycan mimicry by bacteria. Finally, we provide an overview of several techniques that have emerged from the discipline of chemical glycobiology, which can aid in the study of the composition, variability, interaction and functional role of these mimicking glycans.
Collapse
Affiliation(s)
- Hanna de Jong
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Marc M S M Wösten
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Tom Wennekes
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| |
Collapse
|
12
|
Román-Carrasco P, Hemmer W, Cabezas-Cruz A, Hodžić A, de la Fuente J, Swoboda I. The α-Gal Syndrome and Potential Mechanisms. FRONTIERS IN ALLERGY 2021; 2:783279. [PMID: 35386980 PMCID: PMC8974695 DOI: 10.3389/falgy.2021.783279] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
The α-Gal syndrome is a complex allergic disease characterized by the development of specific IgE antibodies against the carbohydrate galactose-α-1,3-galactose (α-Gal), an oligosaccharide present in cells and tissues of non-primate mammals. Individuals with IgE antibodies to α-Gal suffer from a delayed form of anaphylaxis following red meat consumption. There are several features that make the α-Gal syndrome such a unique allergic disease and distinguish it from other food allergies: (1) symptoms causing IgE antibodies are directed against a carbohydrate moiety, (2) the unusual delay between the consumption of the food and the onset of the symptoms, and (3) the fact that primary sensitization to α-Gal occurs via tick bites. This review takes a closer look at the immune response against α-Gal, in healthy and in α-Gal allergic individuals. Furthermore, the similarities and differences between immune response against α-Gal and against the other important glycan moieties associated with allergies, namely cross-reactive carbohydrate determinants (CCDs), are discussed. Then different mechanisms are discussed that could contribute to the delayed onset of symptoms after consumption of mammalian meat. Moreover, our current knowledge on the role of tick bites in the sensitization process is summarized. The tick saliva has been shown to contain proteins carrying α-Gal, but also bioactive molecules, such as prostaglandin E2, which is capable of stimulating an increased expression of anti-inflammatory cytokines while promoting a decrease in the production of proinflammatory mediators. Together these components might promote Th2-related immunity and trigger a class switch to IgE antibodies directed against the oligosaccharide α-Gal. The review also points to open research questions that remain to be answered and proposes future research directions, which will help to get a better understanding and lead to a better management of the disease.
Collapse
Affiliation(s)
- Patricia Román-Carrasco
- Molecular Biotechnology Section, FH Campus Wien, University of Applied Sciences, Vienna, Austria
| | | | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Adnan Hodžić
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - José de la Fuente
- SaBio, Instituto de Investigación de Recursos Cinegéticos, IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Ines Swoboda
- Molecular Biotechnology Section, FH Campus Wien, University of Applied Sciences, Vienna, Austria
| |
Collapse
|
13
|
Kwoji ID, Aiyegoro OA, Okpeku M, Adeleke MA. Multi-Strain Probiotics: Synergy among Isolates Enhances Biological Activities. BIOLOGY 2021; 10:322. [PMID: 33924344 PMCID: PMC8070017 DOI: 10.3390/biology10040322] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
The use of probiotics for health benefits is becoming popular because of the quest for safer products with protective and therapeutic effects against diseases and infectious agents. The emergence and spread of antimicrobial resistance among pathogens had prompted restrictions over the non-therapeutic use of antibiotics for prophylaxis and growth promotion, especially in animal husbandry. While single-strain probiotics are beneficial to health, multi-strain probiotics might be more helpful because of synergy and additive effects among the individual isolates. This article documents the mechanisms by which multi-strain probiotics exert their effects in managing infectious and non-infectious diseases, inhibiting antibiotic-resistant pathogens and health improvement. The administration of multi-strain probiotics was revealed to effectively alleviate bowel tract conditions, such as irritable bowel syndrome, inhibition of pathogens and modulation of the immune system and gut microbiota. Finally, while most of the current research focuses on comparing the effects of multi-strain and single-strain probiotics, there is a dearth of information on the molecular mechanisms of synergy among multi-strain probiotics isolates. This forms a basis for future research in the development of multi-strain probiotics for enhanced health benefits.
Collapse
Affiliation(s)
- Iliya D. Kwoji
- Discipline of Genetics, School of Life Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4000, South Africa; (I.D.K.); (M.O.)
| | - Olayinka A. Aiyegoro
- Gastrointestinal Microbiology and Biotechnology Unit, Agricultural Research Council-Animal Production, Irene 0062, South Africa;
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom 2520, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4000, South Africa; (I.D.K.); (M.O.)
| | - Matthew A. Adeleke
- Discipline of Genetics, School of Life Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4000, South Africa; (I.D.K.); (M.O.)
| |
Collapse
|
14
|
Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet 2021; 22:71-88. [PMID: 33168968 PMCID: PMC7649713 DOI: 10.1038/s41576-020-00292-x] [Citation(s) in RCA: 598] [Impact Index Per Article: 149.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 12/13/2022]
Abstract
Cell-cell interactions orchestrate organismal development, homeostasis and single-cell functions. When cells do not properly interact or improperly decode molecular messages, disease ensues. Thus, the identification and quantification of intercellular signalling pathways has become a common analysis performed across diverse disciplines. The expansion of protein-protein interaction databases and recent advances in RNA sequencing technologies have enabled routine analyses of intercellular signalling from gene expression measurements of bulk and single-cell data sets. In particular, ligand-receptor pairs can be used to infer intercellular communication from the coordinated expression of their cognate genes. In this Review, we highlight discoveries enabled by analyses of cell-cell interactions from transcriptomic data and review the methods and tools used in this context.
Collapse
Affiliation(s)
- Erick Armingol
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Adam Officer
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Division of Biomedical Informatics, University of California, San Diego, La Jolla, CA, USA
| | - Olivier Harismendy
- Division of Biomedical Informatics, University of California, San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
15
|
Kolmeder CA, de Vos WM. Roadmap to functional characterization of the human intestinal microbiota in its interaction with the host. J Pharm Biomed Anal 2020; 194:113751. [PMID: 33328144 DOI: 10.1016/j.jpba.2020.113751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
It is known for more than 100 years that the intestinal microbes are important for the host's health and the last decade this is being intensely studied with a focus on the mechanistic aspects. Among the fundamental functions of the intestinal microbiome are the priming of the immune system, the production of essential vitamins and the energy harvest from foods. By now, several dozens of diseases, both intestinal and non-intestinal related, have been associated with the intestinal microbiome. Initially, this was based on the description of the composition between groups of different health status or treatment arms based on phylogenetic approaches based on the 16S rRNA gene sequences. This way of analysis has mostly moved to the analysis of all the genes or transcripts of the microbiome i.e. metagenomics and meta-transcriptomics. Differences are regularly found but these have to be taken with caution as we still do not know what the majority of genes of the intestinal microbiome are capable of doing. To circumvent this caveat researchers are studying the proteins and the metabolites of the microbiome and the host via metaproteomics and metabolomics approaches. However, also here the complexity is high and only a fraction of signals obtained with high throughput instruments can be identified and assigned to a known protein or molecule. Therefore, modern microbiome research needs advancement of existing and development of new analytical techniques. The usage of model systems like intestinal organoids where samples can be taken and processed rapidly as well as microfluidics systems may help. This review aims to elucidate what we know about the functionality of the human intestinal microbiome, what technologies are advancing this knowledge, and what innovations are still required to further evolve this actively developing field.
Collapse
Affiliation(s)
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Finland; Laboratory of Microbiology, Wageningen University, the Netherlands
| |
Collapse
|
16
|
Romero-Ben E, Mena Barragán T, García de Dionisio E, Sánchez-Fernández EM, Garcia Fernández JM, Guillén-Mancina E, López-Lázaro M, Khiar N. Mannose-coated polydiacetylene (PDA)-based nanomicelles: synthesis, interaction with concanavalin A and application in the water solubilization and delivery of hydrophobic molecules. J Mater Chem B 2020; 7:5930-5946. [PMID: 31512707 DOI: 10.1039/c9tb01218d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Carbohydrate-lectin interactions are involved in a number of relevant biological events including fertilization, immune response, cell adhesion, tumour cell metastasis, and pathogen infection. Lectins are also tissue specific, making carbohydrates not only promising drug candidates but also excellent low molecular weight ligands for active drug delivery system decorations. In order for these interactions to be effective multivalency is essential, as the interaction of a lectin with its cognate monovalent carbohydrate epitope usually takes place with low affinity. Unlike the covalent approach, supramolecular self-assembly of glyco-monomers mediated by non-covalent forces allows accessing multivalent systems with diverse topology, composition, and assembly dynamics in a single step. In order to fine-tune the size and sugar adaptability of spherical micelles at the nanoscale for an optimal glycoside cluster effect, herein we report the synthesis of mannose-coated static micelles from diacetylene-based mannopyranosyl glycolipids differing in the length of the poly(ethyleneglycol) (PEG) chains and the oxidation state of the anomeric sulfur atom. The reported shot-gun like synthetic approach for the synthesis of dilution-insensitive micelles is based on the ability of diacetylenic-based neoglycolipids to self-assemble into micelles in water and to undergo an easy photopolymerization by a simple irradiation at 254 nm. The affinity of the obtained 6 nanosystems was assessed by enzyme-linked lectin assay (ELLA) using the mannose-specific concanavalin A lectin as a model receptor. Relative binding potency enhancements, compared to methyl α-d-mannopyranoside used as control, from 20-, to 29- to 300-fold on a sugar molar basis were observed for micelles derived from sulfonyl-, sulfinyl- and thioglycoside monomers with a tatraethyleneglycol spacer, respectively, indicative of a significant cluster glycoside effect. Moreover, pMic1 micelles are able to solubilize and slowly liberate lipophilic clinically relevant drugs, and show the enhanced cytotoxic effect of docetaxel toward prostate cancer cells. These findings highlight the potential of mannose-coated photopolymerized micelles pMic1 as an efficient nanovector for active delivery of cytotoxic hydrophobic molecules.
Collapse
Affiliation(s)
- E Romero-Ben
- Asymmetric Synthesis and Functional Nanosystems Group. Instituto de Investigaciones Químicas (IIQ), CSIC and Universidad de Sevilla, C/Américo Vespucio 49, 41092, Seville, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Dierking K, Pita L. Receptors Mediating Host-Microbiota Communication in the Metaorganism: The Invertebrate Perspective. Front Immunol 2020; 11:1251. [PMID: 32612612 PMCID: PMC7308585 DOI: 10.3389/fimmu.2020.01251] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022] Open
Abstract
Multicellular organisms live in close association with a plethora of microorganism, which have a profound effect on multiple host functions. As such, the microbiota and its host form an intimate functional entity, termed the metaorganism or holobiont. But how does the metaorganism communicate? Which receptors recognize microbial signals, mediate the effect of the microbiota on host physiology or regulate microbiota composition and homeostasis? In this review we provide an overview on the function of different receptor classes in animal host-microbiota communication. We put a special focus on invertebrate hosts, including both traditional invertebrate models such as Drosophila melanogaster and Caenorhabditis elegans and “non-model” invertebrates in microbiota research. Finally, we highlight the potential of invertebrate systems in studying mechanism of host-microbiota interactions.
Collapse
Affiliation(s)
- Katja Dierking
- Department of Evolutionary Ecology and Genetics, Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Lucía Pita
- RD3 Marine Symbioses, GEOMAR Helmholtz Centre for Ocean Research, Kiel, Germany
| |
Collapse
|
18
|
Naseri R, Navabi SJ, Samimi Z, Mishra AP, Nigam M, Chandra H, Olatunde A, Tijjani H, Morais-Urano RP, Farzaei MH. Targeting Glycoproteins as a therapeutic strategy for diabetes mellitus and its complications. Daru 2020; 28:333-358. [PMID: 32006343 PMCID: PMC7095136 DOI: 10.1007/s40199-020-00327-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Glycoproteins are organic compounds formed from proteins and carbohydrates, which are found in many parts of the living systems including the cell membranes. Furthermore, impaired metabolism of glycoprotein components plays the main role in the pathogenesis of diabetes mellitus. The aim of this study is to investigate the influence of glycoprotein levels in the treatment of diabetes mellitus. METHODS All relevant papers in the English language were compiled by searching electronic databases, including Scopus, PubMed and Cochrane library. The keywords of glycoprotein, diabetes mellitus, glycan, glycosylation, and inhibitor were searched until January 2019. RESULTS Glycoproteins are pivotal elements in the regulation of cell proliferation, growth, maturation and signaling pathways. Moreover, they are involved in drug binding, drug transportation, efflux of chemicals and stability of therapeutic proteins. These functions, structure, composition, linkages, biosynthesis, significance and biological effects are discussed as related to their use as a therapeutic strategy for the treatment of diabetes mellitus and its complications. CONCLUSIONS The findings revealed several chemical and natural compounds have significant beneficial effects on glycoprotein metabolism. The comprehension of glycoprotein structure and functions are very essential and inevitable to enhance the knowledge of glycoengineering for glycoprotein-based therapeutics as may be required for the treatment of diabetes mellitus and its associated complications. Graphical abstract.
Collapse
Affiliation(s)
- Rozita Naseri
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Jafar Navabi
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, Hemwati Nandan Bahuguna Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand, 246174, India.
| | - Manisha Nigam
- Department of Biochemistry, Hemwati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Harish Chandra
- Department of Microbiology, Gurukul Kangri Vishwavidhyalya, Haridwar, Uttarakhand, 249404, India
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, Nigeria
| | - Habibu Tijjani
- Natural Product Research Laboratory, Department of Biochemistry, Bauchi State University, Gadau, Nigeria
| | - Raquel P Morais-Urano
- Instituto de Química de São Carlos, Universidade de São Paulo, 13560-970, São Carlos, SP, Brasil
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Di Rienzi SC, Britton RA. Adaptation of the Gut Microbiota to Modern Dietary Sugars and Sweeteners. Adv Nutr 2020; 11:616-629. [PMID: 31696209 PMCID: PMC7231582 DOI: 10.1093/advances/nmz118] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/15/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
The consumption of sugar has become central to the Western diet. Cost and health concerns associated with sucrose spurred the development and consumption of other sugars and sweeteners, with the average American consuming 10 times more sugar than 100 y ago. In this review, we discuss how gut microbes are affected by changes in the consumption of sugars and other sweeteners through transcriptional, abundance, and genetic adaptations. We propose that these adaptations result in microbes taking on different metabolic, ecological, and genetic profiles along the intestinal tract. We suggest novel approaches to assess the consequences of these changes on host-microbe interactions to determine the safety of novel sugars and sweeteners.
Collapse
Affiliation(s)
- Sara C Di Rienzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
20
|
Lin B, Qing X, Liao J, Zhuo K. Role of Protein Glycosylation in Host-Pathogen Interaction. Cells 2020; 9:E1022. [PMID: 32326128 PMCID: PMC7226260 DOI: 10.3390/cells9041022] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/11/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Host-pathogen interactions are fundamental to our understanding of infectious diseases. Protein glycosylation is one kind of common post-translational modification, forming glycoproteins and modulating numerous important biological processes. It also occurs in host-pathogen interaction, affecting host resistance or pathogen virulence often because glycans regulate protein conformation, activity, and stability, etc. This review summarizes various roles of different glycoproteins during the interaction, which include: host glycoproteins prevent pathogens as barriers; pathogen glycoproteins promote pathogens to attack host proteins as weapons; pathogens glycosylate proteins of the host to enhance virulence; and hosts sense pathogen glycoproteins to induce resistance. In addition, this review also intends to summarize the roles of lectin (a class of protein entangled with glycoprotein) in host-pathogen interactions, including bacterial adhesins, viral lectins or host lectins. Although these studies show the importance of protein glycosylation in host-pathogen interaction, much remains to be discovered about the interaction mechanism.
Collapse
Affiliation(s)
- Borong Lin
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
| | - Xue Qing
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China;
| | - Jinling Liao
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
- Guangdong Eco-Engineering Polytechnic, Guangzhou 510520, China
| | - Kan Zhuo
- Laboratory of Plant Nematology, South China Agricultural University, Guangzhou 510642, China; (B.L.); (J.L.)
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
21
|
Zhang X, Sheng W, Li K, Rong Y, Wu Q, Meng Q, Kong Y, Chen M. Substrate specificity of the galactokinase from the human gut symbiont Akkermansia muciniphila ATCC BAA-835. Enzyme Microb Technol 2020; 139:109568. [PMID: 32732027 DOI: 10.1016/j.enzmictec.2020.109568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022]
Abstract
Galactokinases, which catalyze the phosphorylation of galactose and possible other monosaccharides, can provide an activated sugar donor to synthesize sugar-containing molecules. In this study, a novel galactokinase from human gut symbiont Akkermansia muciniphila ATCC BAA-835 (GalKAmu) was expressed and characterized. GalKAmu displayed broad substrate tolerance, with catalytic activity towards Gal (100 %), GalN (100 %), GalA (20.2 %), Glc (52.5 %), GlcNAc (15.5 %), Xyl (<5%), ManNAc (58 %), ManF (37.4 %) and l-Glc (80 %). Most interestingly, this was the first GalK isoform which can tolerate ManNAc. Thus, our characterization of GalKAmu broadens the substrate selection of galactokinases.
Collapse
Affiliation(s)
- Xunlian Zhang
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Weihao Sheng
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Kun Li
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Yongheng Rong
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Qizheng Wu
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Qingyun Meng
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
| | - Yun Kong
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China.
| | - Min Chen
- The State Key Laboratory of Microbial Technology, National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China.
| |
Collapse
|
22
|
van Wolferen M, Shajahan A, Heinrich K, Brenzinger S, Black IM, Wagner A, Briegel A, Azadi P, Albers SV. Species-Specific Recognition of Sulfolobales Mediated by UV-Inducible Pili and S-Layer Glycosylation Patterns. mBio 2020; 11:e03014-19. [PMID: 32156822 PMCID: PMC7064770 DOI: 10.1128/mbio.03014-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/21/2020] [Indexed: 01/07/2023] Open
Abstract
The UV-inducible pili system of Sulfolobales (Ups) mediates the formation of species-specific cellular aggregates. Within these aggregates, cells exchange DNA to repair DNA double-strand breaks via homologous recombination. Substitution of the Sulfolobus acidocaldarius pilin subunits UpsA and UpsB with their homologs from Sulfolobus tokodaii showed that these subunits facilitate species-specific aggregation. A region of low conservation within the UpsA homologs is primarily important for this specificity. Aggregation assays in the presence of different sugars showed the importance of N-glycosylation in the recognition process. In addition, the N-glycan decorating the S-layer of S. tokodaii is different from the one of S. acidocaldarius Therefore, each Sulfolobus species seems to have developed a unique UpsA binding pocket and unique N-glycan composition to ensure aggregation and, consequently, also DNA exchange with cells from only the same species, which is essential for DNA repair by homologous recombination.IMPORTANCE Type IV pili can be found on the cell surface of many archaea and bacteria where they play important roles in different processes. The UV-inducible pili system of Sulfolobales (Ups) pili from the crenarchaeal Sulfolobales species are essential in establishing species-specific mating partners, thereby assisting in genome stability. With this work, we show that different Sulfolobus species have specific regions in their Ups pili subunits, which allow them to interact only with cells from the same species. Additionally, different Sulfolobus species have unique surface-layer N-glycosylation patterns. We propose that the unique features of each species allow the recognition of specific mating partners. This knowledge for the first time gives insights into the molecular basis of archaeal self-recognition.
Collapse
Affiliation(s)
- Marleen van Wolferen
- Molecular Biology of Archaea, Institute of Biology II-Microbiology, University of Freiburg, Freiburg, Germany
| | - Asif Shajahan
- Complex Carbohydrate Research Center, The University of Georgia, Athens, Georgia, USA
| | - Kristina Heinrich
- Molecular Biology of Archaea, Institute of Biology II-Microbiology, University of Freiburg, Freiburg, Germany
| | | | - Ian M Black
- Complex Carbohydrate Research Center, The University of Georgia, Athens, Georgia, USA
| | - Alexander Wagner
- Molecular Biology of Archaea, Institute of Biology II-Microbiology, University of Freiburg, Freiburg, Germany
| | - Ariane Briegel
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, Georgia, USA
| | - Sonja-Verena Albers
- Molecular Biology of Archaea, Institute of Biology II-Microbiology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
23
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
24
|
Cytoplasmic glycoengineering enables biosynthesis of nanoscale glycoprotein assemblies. Nat Commun 2019; 10:5403. [PMID: 31776333 PMCID: PMC6881330 DOI: 10.1038/s41467-019-13283-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
Glycosylation of proteins profoundly impacts their physical and biological properties. Yet our ability to engineer novel glycoprotein structures remains limited. Established bacterial glycoengineering platforms require secretion of the acceptor protein to the periplasmic space and preassembly of the oligosaccharide substrate as a lipid-linked precursor, limiting access to protein and glycan substrates respectively. Here, we circumvent these bottlenecks by developing a facile glycoengineering platform that operates in the bacterial cytoplasm. The Glycoli platform leverages a recently discovered site-specific polypeptide glycosyltransferase together with variable glycosyltransferase modules to synthesize defined glycans, of bacterial or mammalian origin, directly onto recombinant proteins in the E. coli cytoplasm. We exploit the cytoplasmic localization of this glycoengineering platform to generate a variety of multivalent glycostructures, including self-assembling nanomaterials bearing hundreds of copies of the glycan epitope. This work establishes cytoplasmic glycoengineering as a powerful platform for producing glycoprotein structures with diverse future biomedical applications. Established bacterial glycoengineering platforms limit access to protein and glycan substrates. Here the authors design a cytoplasmic protein glycosylation system, Glycoli, to generate a variety of multivalent glycostructures.
Collapse
|
25
|
Expansion of the Spore Surface Polysaccharide Layer in Bacillus subtilis by Deletion of Genes Encoding Glycosyltransferases and Glucose Modification Enzymes. J Bacteriol 2019; 201:JB.00321-19. [PMID: 31235516 DOI: 10.1128/jb.00321-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022] Open
Abstract
Polysaccharides (PS) decorate the surface of dormant endospores (spores). In the model organism for sporulation, Bacillus subtilis, the composition of the spore PS is not known in detail. Here, we have assessed how PS synthesis enzymes produced during the late stages of sporulation affect spore surface properties. Using four methods, bacterial adhesion to hydrocarbons (BATH) assays, India ink staining, transmission electron microscopy (TEM) with ruthenium red staining, and scanning electron microscopy (SEM), we characterized the contributions of four sporulation gene clusters, spsABCDEFGHIJKL, yfnHGF-yfnED, ytdA-ytcABC, and cgeAB-cgeCDE, on the morphology and properties of the crust, the outermost spore layer. Our results show that all mutations in the sps operon result in the production of spores that are more hydrophobic and lack a visible crust, presumably because of reduced PS deposition, while mutations in cgeD and the yfnH-D cluster noticeably expand the PS layer. In addition, yfnH-D mutant spores exhibit a crust with an unusual weblike morphology. The hydrophobic phenotype from sps mutant spores was partially rescued by a second mutation inactivating any gene in the yfnHGF operon. While spsI, yfnH, and ytdA are paralogous genes, all encoding glucose-1-phosphate nucleotidyltransferases, each paralog appears to contribute in a distinct manner to the spore PS. Our data are consistent with the possibility that each gene cluster is responsible for the production of its own respective deoxyhexose. In summary, we found that disruptions to the PS layer modify spore surface hydrophobicity and that there are multiple saccharide synthesis pathways involved in spore surface properties.IMPORTANCE Many bacteria are characterized by their ability to form highly resistant spores. The dormant spore state allows these species to survive even the harshest treatments with antimicrobial agents. Spore surface properties are particularly relevant because they influence spore dispersal in various habitats from natural to human-made environments. The spore surface in Bacillus subtilis (crust) is composed of a combination of proteins and polysaccharides. By inactivating the enzymes responsible for the synthesis of spore polysaccharides, we can assess how spore surface properties such as hydrophobicity are modulated by the addition of specific carbohydrates. Our findings indicate that several sporulation gene clusters are responsible for the assembly and allocation of surface polysaccharides. Similar mechanisms could be modulating the dispersal of infectious spore-forming bacteria.
Collapse
|
26
|
Uprety T, Spurlin BB, Antony L, Sreenivasan C, Young A, Li F, Hildreth MB, Kaushik RS. Development and characterization of a stable bovine intestinal sub-epithelial myofibroblast cell line from ileum of a young calf. In Vitro Cell Dev Biol Anim 2019; 55:533-547. [PMID: 31183683 DOI: 10.1007/s11626-019-00365-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
Intestinal sub-epithelial myofibroblasts (ISEMFs) are mesenchymal cells that do not express cytokeratin but express α-smooth muscle actin and vimentin. Despite being cells with diverse functions, there is a paucity of knowledge about their origin and functions primarily due to the absence of a stable cell line. Although myofibroblast in vitro models for human, mouse, and pig are available, there is no ISEMF cell line available from young calves. We isolated and developed an ileal ISEMF cell line from a 2-d-old calf that expressed α-smooth muscle actin and vimentin but no cytokeratin indicating true myofibroblast cells. To overcome replicative senescence, we immortalized primary cells with SV40 large T antigen. We characterized and compared both primary and immortalized ileal ISEMF cells for surface glycan and Toll-like-receptor (TLR) expression by lectin-binding assay and real-time quantitative PCR (RT-qPCR) assay respectively. SV40 immortalization significantly decreased surface lectin binding for lectins GSL-I, PHA-L, ECL, Jacalin, Con-A, LCA, and LEL. Both cell types expressed TLRs 1-9 and showed no significant differences in TLR expression. Thus, these cells can be useful in vitro model to study ISEMF's origin, physiology, and functions.
Collapse
Affiliation(s)
- Tirth Uprety
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Brionna B Spurlin
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Linto Antony
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Chithra Sreenivasan
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Alan Young
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
| | - Feng Li
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Michael B Hildreth
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA
| | - Radhey S Kaushik
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, 57007, USA.
| |
Collapse
|
27
|
van der Veer C, Hertzberger RY, Bruisten SM, Tytgat HLP, Swanenburg J, de Kat Angelino-Bart A, Schuren F, Molenaar D, Reid G, de Vries H, Kort R. Comparative genomics of human Lactobacillus crispatus isolates reveals genes for glycosylation and glycogen degradation: implications for in vivo dominance of the vaginal microbiota. MICROBIOME 2019; 7:49. [PMID: 30925932 PMCID: PMC6441167 DOI: 10.1186/s40168-019-0667-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/15/2019] [Indexed: 05/17/2023]
Abstract
BACKGROUND A vaginal microbiota dominated by lactobacilli (particularly Lactobacillus crispatus) is associated with vaginal health, whereas a vaginal microbiota not dominated by lactobacilli is considered dysbiotic. Here we investigated whether L. crispatus strains isolated from the vaginal tract of women with Lactobacillus-dominated vaginal microbiota (LVM) are pheno- or genotypically distinct from L. crispatus strains isolated from vaginal samples with dysbiotic vaginal microbiota (DVM). RESULTS We studied 33 L. crispatus strains (n = 16 from LVM; n = 17 from DVM). Comparison of these two groups of strains showed that, although strain differences existed, both groups degraded various carbohydrates, produced similar amounts of organic acids, inhibited Neisseria gonorrhoeae growth, and did not produce biofilms. Comparative genomics analyses of 28 strains (n = 12 LVM; n = 16 DVM) revealed a novel, 3-fragmented glycosyltransferase gene that was more prevalent among strains isolated from DVM. Most L. crispatus strains showed growth on glycogen-supplemented growth media. Strains that showed less-efficient (n = 6) or no (n = 1) growth on glycogen all carried N-terminal deletions (respectively, 29 and 37 amino acid deletions) in a putative pullulanase type I protein. DISCUSSION L. crispatus strains isolated from LVM were not phenotypically distinct from L. crispatus strains isolated from DVM; however, the finding that the latter were more likely to carry a 3-fragmented glycosyltransferase gene may indicate a role for cell surface glycoconjugates, which may shape vaginal microbiota-host interactions. Furthermore, the observation that variation in the pullulanase type I gene is associated with growth on glycogen discourages previous claims that L. crispatus cannot directly utilize glycogen.
Collapse
Affiliation(s)
- Charlotte van der Veer
- Department of Infectious Diseases, Public Health Service, GGD, Amsterdam, The Netherlands
| | - Rosanne Y Hertzberger
- Department of Molecular Cell Biology, Faculty of Science, O|2 Lab Building, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Sylvia M Bruisten
- Department of Infectious Diseases, Public Health Service, GGD, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Jorne Swanenburg
- Department of Molecular Cell Biology, Faculty of Science, O|2 Lab Building, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Netherlands Organization for Applied Scientific Research (TNO), Microbiology and Systems Biology, Zeist, The Netherlands
| | - Alie de Kat Angelino-Bart
- Netherlands Organization for Applied Scientific Research (TNO), Microbiology and Systems Biology, Zeist, The Netherlands
| | - Frank Schuren
- Netherlands Organization for Applied Scientific Research (TNO), Microbiology and Systems Biology, Zeist, The Netherlands
| | - Douwe Molenaar
- Department of Molecular Cell Biology, Faculty of Science, O|2 Lab Building, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Gregor Reid
- Canadian R&D Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, Canada
- Departments of Microbiology and Immunology, and Surgery, Western University, London, Ontario, Canada
| | - Henry de Vries
- Department of Infectious Diseases, Public Health Service, GGD, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Remco Kort
- Department of Molecular Cell Biology, Faculty of Science, O|2 Lab Building, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Netherlands Organization for Applied Scientific Research (TNO), Microbiology and Systems Biology, Zeist, The Netherlands.
- ARTIS-Micropia, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Latousakis D, Nepravishta R, Rejzek M, Wegmann U, Le Gall G, Kavanaugh D, Colquhoun IJ, Frese S, MacKenzie DA, Walter J, Angulo J, Field RA, Juge N. Serine-rich repeat protein adhesins from Lactobacillus reuteri display strain specific glycosylation profiles. Glycobiology 2019; 29:45-58. [PMID: 30371779 PMCID: PMC6291802 DOI: 10.1093/glycob/cwy100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/19/2018] [Accepted: 10/25/2018] [Indexed: 01/24/2023] Open
Abstract
Lactobacillus reuteri is a gut symbiont inhabiting the gastrointestinal tract of numerous vertebrates. The surface-exposed serine-rich repeat protein (SRRP) is a major adhesin in Gram-positive bacteria. Using lectin and sugar nucleotide profiling of wild-type or L. reuteri isogenic mutants, MALDI-ToF-MS, LC-MS and GC-MS analyses of SRRPs, we showed that L. reuteri strains 100-23C (from rodent) and ATCC 53608 (from pig) can perform protein O-glycosylation and modify SRRP100-23 and SRRP53608 with Hex-Glc-GlcNAc and di-GlcNAc moieties, respectively. Furthermore, in vivo glycoengineering in E. coli led to glycosylation of SRRP53608 variants with α-GlcNAc and GlcNAcβ(1→6)GlcNAcα moieties. The glycosyltransferases involved in the modification of these adhesins were identified within the SecA2/Y2 accessory secretion system and their sugar nucleotide preference determined by saturation transfer difference NMR spectroscopy and differential scanning fluorimetry. Together, these findings provide novel insights into the cellular O-protein glycosylation pathways of gut commensal bacteria and potential routes for glycoengineering applications.
Collapse
Affiliation(s)
- Dimitrios Latousakis
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Ridvan Nepravishta
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Martin Rejzek
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, UK
| | - Udo Wegmann
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Gwenaelle Le Gall
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Devon Kavanaugh
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Ian J Colquhoun
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Donald A MacKenzie
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Jesus Angulo
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Robert A Field
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, UK
| | - Nathalie Juge
- The Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| |
Collapse
|
29
|
Abstract
Glycosylation is one of the most prevalent posttranslational modifications that profoundly affects the structure and functions of proteins in a wide variety of biological recognition events. However, the structural complexity and heterogeneity of glycoproteins, usually resulting from the variations of glycan components and/or the sites of glycosylation, often complicates detailed structure-function relationship studies and hampers the therapeutic applications of glycoproteins. To address these challenges, various chemical and biological strategies have been developed for producing glycan-defined homogeneous glycoproteins. This review highlights recent advances in the development of chemoenzymatic methods for synthesizing homogeneous glycoproteins, including the generation of various glycosynthases for synthetic purposes, endoglycosidase-catalyzed glycoprotein synthesis and glycan remodeling, and direct enzymatic glycosylation of polypeptides and proteins. The scope, limitation, and future directions of each method are discussed.
Collapse
Affiliation(s)
- Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
30
|
Tytgat HLP, Nobrega FL, van der Oost J, de Vos WM. Bowel Biofilms: Tipping Points between a Healthy and Compromised Gut? Trends Microbiol 2018; 27:17-25. [PMID: 30219265 DOI: 10.1016/j.tim.2018.08.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/29/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023]
Abstract
Bacterial communities are known to impact human health and disease. Mixed species biofilms, mostly pathogenic in nature, have been observed in dental and gastric infections as well as in intestinal diseases, chronic gut wounds and colon cancer. Apart from the appendix, the presence of thick polymicrobial biofilms in the healthy gut mucosa is still debated. Polymicrobial biofilms containing potential pathogens appear to be an early-warning signal of developing disease and can be regarded as a tipping point between a healthy and a diseased state of the gut mucosa. Key biofilm-forming pathogens and associated molecules hold promise as biomarkers. Criteria to distinguish microcolonies from biofilms are crucial to provide clarity when reporting biofilm-related phenomena in health and disease in the gut.
Collapse
Affiliation(s)
- Hanne L P Tytgat
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands; Institute of Microbiology, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Franklin L Nobrega
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands; Kavli Institute of Nanoscience and Department of BioNanoscience, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - John van der Oost
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands; Faculty of Medicine, Immunobiology Research Program, Department of Bacteriology and Immunology, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
31
|
van Beusekom B, Lütteke T, Joosten RP. Making glycoproteins a little bit sweeter with PDB-REDO. Acta Crystallogr F Struct Biol Commun 2018; 74:463-472. [PMID: 30084395 PMCID: PMC6096482 DOI: 10.1107/s2053230x18004016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/07/2018] [Indexed: 02/04/2023] Open
Abstract
Glycosylation is one of the most common forms of protein post-translational modification, but is also the most complex. Dealing with glycoproteins in structure model building, refinement, validation and PDB deposition is more error-prone than dealing with nonglycosylated proteins owing to limitations of the experimental data and available software tools. Also, experimentalists are typically less experienced in dealing with carbohydrate residues than with amino-acid residues. The results of the reannotation and re-refinement by PDB-REDO of 8114 glycoprotein structure models from the Protein Data Bank are analyzed. The positive aspects of 3620 reannotations and subsequent refinement, as well as the remaining challenges to obtaining consistently high-quality carbohydrate models, are discussed.
Collapse
Affiliation(s)
- Bart van Beusekom
- Division of Biochemistry, Netherlands Cancer Insitute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Thomas Lütteke
- Institute of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
| | - Robbie P. Joosten
- Division of Biochemistry, Netherlands Cancer Insitute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
32
|
Formosa-Dague C, Castelain M, Martin-Yken H, Dunker K, Dague E, Sletmoen M. The Role of Glycans in Bacterial Adhesion to Mucosal Surfaces: How Can Single-Molecule Techniques Advance Our Understanding? Microorganisms 2018; 6:E39. [PMID: 29734645 PMCID: PMC6027152 DOI: 10.3390/microorganisms6020039] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022] Open
Abstract
Bacterial adhesion is currently the subject of increased interest from the research community, leading to fast progress in our understanding of this complex phenomenon. Resent research within this field has documented the important roles played by glycans for bacterial surface adhesion, either through interaction with lectins or with other glycans. In parallel with this increased interest for and understanding of bacterial adhesion, there has been a growth in the sophistication and use of sensitive force probes for single-molecule and single cell studies. In this review, we highlight how the sensitive force probes atomic force microscopy (AFM) and optical tweezers (OT) have contributed to clarifying the mechanisms underlying bacterial adhesion to glycosylated surfaces in general and mucosal surfaces in particular. We also describe research areas where these techniques have not yet been applied, but where their capabilities appear appropriate to advance our understanding.
Collapse
Affiliation(s)
| | - Mickaël Castelain
- LISBP, Université de Toulouse, CNRS, INRA, INSA, 31400 Toulouse, France.
| | - Hélène Martin-Yken
- LISBP, Université de Toulouse, CNRS, INRA, INSA, 31400 Toulouse, France.
| | - Karen Dunker
- Department of Biotechnology and Food Science, NTNU the Norwegian University of Science and Technology, NO-7491 Trondheim, Norway.
| | - Etienne Dague
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France.
| | - Marit Sletmoen
- Department of Biotechnology and Food Science, NTNU the Norwegian University of Science and Technology, NO-7491 Trondheim, Norway.
| |
Collapse
|
33
|
Structural basis for the role of serine-rich repeat proteins from Lactobacillus reuteri in gut microbe-host interactions. Proc Natl Acad Sci U S A 2018; 115:E2706-E2715. [PMID: 29507249 PMCID: PMC5866549 DOI: 10.1073/pnas.1715016115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Gut bacteria play a key role in health and disease, but the molecular mechanisms underpinning their interaction with the host remain elusive. The serine-rich repeat proteins (SRRPs) are a family of adhesins identified in many Gram-positive pathogenic bacteria. We previously showed that beneficial bacterial species found in the gut also express SRRPs and that SRRP was required for the ability of Lactobacillus reuteri strain to colonize mice. Here, our structural and biochemical data reveal that L. reuteri SRRP adopts a β-solenoid fold not observed in other structurally characterized SRRPs and functions as an adhesin via a pH-dependent mechanism, providing structural insights into the role of these adhesins in biofilm formation of gut symbionts. Lactobacillus reuteri, a Gram-positive bacterial species inhabiting the gastrointestinal tract of vertebrates, displays remarkable host adaptation. Previous mutational analyses of rodent strain L. reuteri 100-23C identified a gene encoding a predicted surface-exposed serine-rich repeat protein (SRRP100-23) that was vital for L. reuteri biofilm formation in mice. SRRPs have emerged as an important group of surface proteins on many pathogens, but no structural information is available in commensal bacteria. Here we report the 2.00-Å and 1.92-Å crystal structures of the binding regions (BRs) of SRRP100-23 and SRRP53608 from L. reuteri ATCC 53608, revealing a unique β-solenoid fold in this important adhesin family. SRRP53608-BR bound to host epithelial cells and DNA at neutral pH and recognized polygalacturonic acid (PGA), rhamnogalacturonan I, or chondroitin sulfate A at acidic pH. Mutagenesis confirmed the role of the BR putative binding site in the interaction of SRRP53608-BR with PGA. Long molecular dynamics simulations showed that SRRP53608-BR undergoes a pH-dependent conformational change. Together, these findings provide mechanistic insights into the role of SRRPs in host–microbe interactions and open avenues of research into the use of biofilm-forming probiotics against clinically important pathogens.
Collapse
|
34
|
Douillard FP, Mora D, Eijlander RT, Wels M, de Vos WM. Comparative genomic analysis of the multispecies probiotic-marketed product VSL#3. PLoS One 2018; 13:e0192452. [PMID: 29451876 PMCID: PMC5815585 DOI: 10.1371/journal.pone.0192452] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Several probiotic-marketed formulations available for the consumers contain live lactic acid bacteria and/or bifidobacteria. The multispecies product commercialized as VSL#3 has been used for treating various gastro-intestinal disorders. However, like many other products, the bacterial strains present in VSL#3 have only been characterized to a limited extent and their efficacy as well as their predicted mode of action remain unclear, preventing further applications or comparative studies. In this work, the genomes of all eight bacterial strains present in VSL#3 were sequenced and characterized, to advance insights into the possible mode of action of this product and also to serve as a basis for future work and trials. Phylogenetic and genomic data analysis allowed us to identify the 7 species present in the VSL#3 product as specified by the manufacturer. The 8 strains present belong to the species Streptococcus thermophilus, Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus plantarum, Lactobacillus helveticus, Bifidobacterium breve and B. animalis subsp. lactis (two distinct strains). Comparative genomics revealed that the draft genomes of the S. thermophilus and L. helveticus strains were predicted to encode most of the defence systems such as restriction modification and CRISPR-Cas systems. Genes associated with a variety of potential probiotic functions were also identified. Thus, in the three Bifidobacterium spp., gene clusters were predicted to encode tight adherence pili, known to promote bacteria-host interaction and intestinal barrier integrity, and to impact host cell development. Various repertoires of putative signalling proteins were predicted to be encoded by the genomes of the Lactobacillus spp., i.e. surface layer proteins, LPXTG-containing proteins, or sortase-dependent pili that may interact with the intestinal mucosa and dendritic cells. Taken altogether, the individual genomic characterization of the strains present in the VSL#3 product confirmed the product specifications, determined its coding capacity as well as identified potential probiotic functions.
Collapse
Affiliation(s)
- François P. Douillard
- Research Program Unit Immunobiology, Medicum, University of Helsinki, Helsinki, Finland
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Diego Mora
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | | | | | - Willem M. de Vos
- Research Program Unit Immunobiology, Medicum, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- * E-mail:
| |
Collapse
|
35
|
Powers MJ, Trent MS. Expanding the paradigm for the outer membrane: Acinetobacter baumannii in the absence of endotoxin. Mol Microbiol 2017; 107:47-56. [PMID: 29114953 DOI: 10.1111/mmi.13872] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 12/30/2022]
Abstract
Asymmetry in the outer membrane has long defined the cell envelope of Gram-negative bacteria. This asymmetry, with lipopolysaccharide (LPS) or lipooligosaccharide (LOS) exclusively in the outer leaflet of the membrane, establishes an impermeable barrier that protects the cell from a number of stressors in the environment. Work done over the past 5 years has shown that Acinetobacter baumannii has the remarkable capability to survive with inactivated production of lipid A biosynthesis and the absence of LOS in its outer membrane. The implications of LOS-deficient A. baumannii are far-reaching - from impacts on cell envelope biogenesis and maintenance, bacterial physiology, antibiotic resistance and virulence. This review examines recent work that has contributed to our understanding of LOS-deficiency and compares it to studies done on Neisseria meningitidis and Moraxella catarrhalis; the two other organisms with this capability.
Collapse
Affiliation(s)
- Matthew Joseph Powers
- Department of Infectious Diseases, University of Georgia, 510 DW Brooks Drive, Athens, GA 30602, USA.,Department of Microbiology, University of Georgia, Athens, GA, USA
| | - M Stephen Trent
- Department of Infectious Diseases, University of Georgia, 510 DW Brooks Drive, Athens, GA 30602, USA
| |
Collapse
|
36
|
|
37
|
Jaroentomeechai T, Zheng X, Hershewe J, Stark JC, Jewett MC, DeLisa MP. A Pipeline for Studying and Engineering Single-Subunit Oligosaccharyltransferases. Methods Enzymol 2017; 597:55-81. [PMID: 28935112 DOI: 10.1016/bs.mie.2017.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Asparagine-linked (N-linked) protein glycosylation is one of the most abundant types of posttranslational modification, occurring in all domains of life. The central enzyme in N-linked glycosylation is the oligosaccharyltransferase (OST), which catalyzes the covalent attachment of preassembled glycans to specific asparagine residues in target proteins. Whereas in higher eukaryotes the OST is comprised of eight different membrane proteins, of which the catalytic subunit is STT3, in kinetoplastids and prokaryotes the OST is a monomeric enzyme bearing homology to STT3. Given their relative simplicity, these single-subunit OSTs (ssOSTs) have emerged as important targets for mechanistic dissection of poorly understood aspects of N-glycosylation and at the same time hold great potential for the biosynthesis of custom glycoproteins. To take advantage of this utility, this chapter describes a multipronged approach for studying and engineering ssOSTs that integrates in vivo screening technology with in vitro characterization methods, thereby creating a versatile and readily adaptable pipeline for virtually any ssOST of interest.
Collapse
Affiliation(s)
- Thapakorn Jaroentomeechai
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | - Xiaolu Zheng
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
| | | | | | - Michael C Jewett
- Northwestern University, Evanston, IL, United States; Center for Synthetic Biology, Northwestern University, Evanston, IL, United States
| | - Matthew P DeLisa
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
38
|
Valeriano VDV, Balolong MP, Kang DK. Probiotic roles of Lactobacillus sp. in swine: insights from gut microbiota. J Appl Microbiol 2017; 122:554-567. [PMID: 27914202 DOI: 10.1111/jam.13364] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/11/2016] [Accepted: 11/24/2016] [Indexed: 12/13/2022]
Abstract
The use of lactobacilli as probiotics in swine has been gaining attention due to their ability to improve growth performance and carcass quality, prevent gastrointestinal infection and most importantly, their 'generally recognized as safe' status. Previous studies support the potential of lactobacilli to regulate host immune systems, enhance gut metabolic capacities and maintain balance in the gut microbiota. Research on swine gut microbiota has revealed complex gut microbial community structure and showed the importance of Lactobacillus to the host's health. However, the species- and strain-specific characteristics of lactobacilli that confer probiotic benefits are still not well understood. The diversity of probiotic traits in a complex gut ecosystem makes it challenging to infer the relationships between specific functions of Lactobacillus sp. and host health. In this review, we provide an overview of how lactobacilli play a pivotal role in the swine gut ecosystem and identify key characteristics that influence gut microbial community structure and the health of pigs. In addition, based on recent and ongoing meta-omics and omics research on the gut microbiota of pigs, we suggest a workflow combining culture-dependent and culture-independent approaches for more effective selection of probiotic lactobacilli.
Collapse
Affiliation(s)
- V D V Valeriano
- Department of Animal Resources Science, Dankook University, Cheonan, Korea
| | - M P Balolong
- Department of Animal Resources Science, Dankook University, Cheonan, Korea.,Department of Biology, College of Arts and Sciences, University of the Philippines, Manila, Philippines
| | - D-K Kang
- Department of Animal Resources Science, Dankook University, Cheonan, Korea
| |
Collapse
|
39
|
Abstract
The idea you could use lactic acid bacteria to treat and prevent recurrence of vaginal infections was ridiculed in the early 1980s. Bacteria were the bad guys to be eradicated by current and emerging antibiotic classes. Thirty years later, probiotic administration of microbes is widespread worldwide, including for vaginal and bladder health in women, and the scientific basis and clinical efficacy data for this and multiple other applications prove the viability of this concept. The development of this approach, the creation of a definition for probiotics, and the expansion to other areas of women’s health form the basis of this review.
Collapse
Affiliation(s)
- Gregor Reid
- Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2, Canada
- Departments of Surgery and Microbiology and Immunology, University of Western Ontario, Richmond Street, London, ON N6A 3K7, Canada
| |
Collapse
|
40
|
Barel M, Charbit A. Role of Glycosylation/Deglycolysation Processes in Francisella tularensis Pathogenesis. Front Cell Infect Microbiol 2017; 7:71. [PMID: 28377902 PMCID: PMC5359314 DOI: 10.3389/fcimb.2017.00071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis is able to invade, survive and replicate inside a variety of cell types. However, in vivo F. tularensis preferentially enters host macrophages where it rapidly escapes to the cytosol to avoid phagosomal stresses and to multiply to high numbers. We previously showed that human monocyte infection by F. tularensis LVS triggered deglycosylation of the glutamine transporter SLC1A5. However, this deglycosylation, specifically induced by Francisella infection, was not restricted to SLC1A5, suggesting that host protein deglycosylation processes in general might contribute to intracellular bacterial adaptation. Indeed, we later found that Francisella infection modulated the transcription of numerous glycosidase and glycosyltransferase genes in human macrophages and analysis of cell extracts revealed an important increase of N and O-protein glycosylation. In eukaryotic cells, glycosylation has significant effects on protein folding, conformation, distribution, stability, and activity and dysfunction of protein glycosylation may lead to development of diseases like cancer and pathogenesis of infectious diseases. Pathogenic bacteria have also evolved dedicated glycosylation machineries and have notably been shown to use these glycoconjugates as ligands to specifically interact with the host. In this review, we will focus on Francisella and summarize our current understanding of the importance of these post-translational modifications on its intracellular niche adaptation.
Collapse
Affiliation(s)
- Monique Barel
- Sorbonne Paris Cité, Bâtiment Leriche, Université Paris DescartesParis, France; Institut National de la Santé et de la Recherche Médicale, Institut Necker-Enfants Malades, INSERM U1151 -Team 11, Pathogenesis of Systemic InfectionsParis, France; Centre National de la Recherche Scientifique, UMR8253Paris, France
| | - Alain Charbit
- Sorbonne Paris Cité, Bâtiment Leriche, Université Paris DescartesParis, France; Institut National de la Santé et de la Recherche Médicale, Institut Necker-Enfants Malades, INSERM U1151 -Team 11, Pathogenesis of Systemic InfectionsParis, France; Centre National de la Recherche Scientifique, UMR8253Paris, France
| |
Collapse
|
41
|
Affiliation(s)
- Stefan Gaunitz
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Gabe Nagy
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Nicola L. B. Pohl
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Milos V. Novotny
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
- Regional Center for Applied Molecular Oncology, Masaryk Memorial Oncological Institute, 656 53 Brno, Czech Republic
| |
Collapse
|