1
|
Ndeh DA, Nakjang S, Kwiatkowski KJ, Sawyers C, Koropatkin NM, Hirt RP, Bolam DN. A Bacteroides thetaiotaomicron genetic locus encodes activities consistent with mucin O-glycoprotein processing and N-acetylgalactosamine metabolism. Nat Commun 2025; 16:3485. [PMID: 40216766 PMCID: PMC11992087 DOI: 10.1038/s41467-025-58660-2] [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: 01/17/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The gut microbiota is a key modulator of human health and the status of major diseases including cancer, diabetes and inflammatory bowel disease. Central to microbiota survival is the ability to metabolise complex dietary and host-derived glycans, including intestinal mucins. The prominent human gut microbe Bacteroides thetaiotaomicron (B. theta) is a versatile and highly efficient complex glycan degrader thanks to the expansion of gene clusters termed polysaccharide utilisation loci (PULs). While the mechanism of action for several singular dietary glycan-induced PULs have been elucidated, studies on the unusually high number of mucin-inducible PULs in B. theta significantly lag behind. Here we show that a mucin inducible PUL BT4240-50 encodes activities consistent with the processing and metabolism of mucin O-glycoproteins and their core sugar N-acetylgalactosamine (GalNAc). PUL BT4240-50 was also shown to be important for competitive growth on mucins in vitro, encoding a kinase (BT4240) critical for GalNAc metabolism. Additionally, BT4240-kinase was shown to be essential for glycosaminoglycan metabolism, extending the PULs function beyond mucins. These data advance our understanding of glycoprotein metabolism at mucosal surfaces, highlighting GalNAc as a key metabolite for competitive microbial survival in the human gut.
Collapse
Affiliation(s)
- Didier A Ndeh
- Division of Plant Sciences, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Sirintra Nakjang
- Precision Medicine Centre of Excellence, Queen's University Belfast, Belfast, UK
| | - Kurt J Kwiatkowski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Claire Sawyers
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Nicole M Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert P Hirt
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - David N Bolam
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
2
|
Boccacci Y, Dumont N, Doyon Y, Laganière J. CRISPR-Cas9-driven antigen conversion of clinically relevant blood group systems. Hum Mol Genet 2025:ddaf040. [PMID: 40172074 DOI: 10.1093/hmg/ddaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025] Open
Abstract
The common practice of blood transfusion entirely relies on blood donations from the population. Ensuring blood group compatibility between a donor and a recipient is paramount to prevent critical adverse reactions. Finding compatible blood can be challenging given the high diversity of blood group antigens, especially for chronically transfused patients at higher risk of alloimmunization owing to repeated exposures to foreign RBCs. In addition, due to the immunogenicity of the ABO blood group and the highly polymorphic nature of the Rhesus (Rh) system, they both remain of prime importance in transfusion medicine. Cultured red blood cells (cRBCs) may eventually provide an alternative for blood donations-at least in some circumstances. Combining cRBCs with blood group gene editing could broaden transfusion accessibility by making antigen expression compatible with rare phenotypes, thus meeting the needs of more patients. Starting from mobilized, erythroid-primed hematopoietic stem and progenitor cells (HSPCs), we used virus- and selection-free, CRISPR-Cas9-mediated knockouts to produce erythroid cells devoid of AB and Rh antigen. The approach yielded almost complete conversion to O- and RhNull phenotypes, as determined by standard hemagglutination and flow cytometry analyses. Combined with robust cRBC protocols, these clinically relevant phenotypic changes could eventually expand the accessibility of blood transfusion for specific and unmet clinical needs.
Collapse
Affiliation(s)
- Yelena Boccacci
- Centre Hospitalier Universitaire de Québec Research Center - Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
- Université Laval Cancer Research Centre, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
- Medical Affairs and Innovation, Héma-Québec, 1070 avenue des Sciences-de-la-Vie, Québec, QC G1V 5C3, Canada
| | - Nellie Dumont
- Medical Affairs and Innovation, Héma-Québec, 1070 avenue des Sciences-de-la-Vie, Québec, QC G1V 5C3, Canada
| | - Yannick Doyon
- Centre Hospitalier Universitaire de Québec Research Center - Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
- Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
- Université Laval Cancer Research Centre, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Josée Laganière
- Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
- Medical Affairs and Innovation, Héma-Québec, 1070 avenue des Sciences-de-la-Vie, Québec, QC G1V 5C3, Canada
| |
Collapse
|
3
|
Möller C, Terholsen H, Schmöker O, Lê TLA, Wesche J, Schmiade P, Eppendorfer E, Rimkus N, Girbardt B, Böttcher D, Palm GJ, Hoppen J, Lammers M, Greinacher A, Aurich K, Bornscheuer UT. Identification and Protein Engineering of Galactosidases for the Conversion of Blood Type B to Blood Type O. Chembiochem 2025; 26:e202500072. [PMID: 40013737 PMCID: PMC12002104 DOI: 10.1002/cbic.202500072] [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: 01/25/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 02/28/2025]
Abstract
The supply of blood products such as red blood cells poses a challenge due to rising demand and declining donor numbers. Careful matching of blood products of different types is required. Only type O of the blood types A, B, AB and O can be received by any patient without transfusion incompatibilities. Therefore, O-type blood can be considered "universal blood" and is especially needed in emergency situations. In this study, we focused on the conversion of the B antigen by enzymatic deglycosylation to generate the H antigen determining O-type blood. For this, we characterized several previously unstudied α-1,3-galactosidases belonging to the GH110 family. Our findings revealed that the α-1,3-galactosidase from Pedobacter panaciterrae (PpaGal) exhibits superior efficiency compared to previously described galactosidases. We further increased the activity of PpaGal by 2.5-fold using site-directed mutagenesis. Moreover, we solved two crystal structures of PpaGal, one in the apo-state and another in complex with d-galactose. The combination of our mutagenesis study with the solved crystal structures provides valuable information to guide further optimization of PpaGal or other B antigen converting enzymes paving the way for the easier production of universal blood from B-type blood.
Collapse
Affiliation(s)
- Christina Möller
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Henrik Terholsen
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Ole Schmöker
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Thi Linh Anne Lê
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Jan Wesche
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Paula Schmiade
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Esther Eppendorfer
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Niklas Rimkus
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Britta Girbardt
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Dominique Böttcher
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Gottfried J. Palm
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Jens Hoppen
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Michael Lammers
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Andreas Greinacher
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Konstanze Aurich
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Uwe T. Bornscheuer
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| |
Collapse
|
4
|
Zeng J, Ma M, Jiang X, Rao Z, Huang D, Zhang H, Yin S, Bao R, Zhang H, Wang Z, Gao H, Gong F, Lin T, Zhang K, Song T. Enzymatic conversion of blood group B kidney prevents hyperacute antibody-mediated injuries in ABO-incompatible transplantation. Nat Commun 2025; 16:1506. [PMID: 39929829 PMCID: PMC11810989 DOI: 10.1038/s41467-025-56563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Matching ABO blood group antigens between donors and recipients is critical to prevent hyperacute rejection in kidney transplantation. Enzymatic conversion of blood group antigens to the universal O type presents a promising strategy to overcome barriers in ABO-incompatible kidney transplantation. In this study, we employ α-galactosidase from Bacteroides fragilis to convert type B kidneys to type O during hypothermic machine perfusion. After 3 hours of perfusion with enzyme, more than 95% of blood group B antigens in the kidney endothelium are effectively removed. Subsequently, enzyme-treated kidneys are protected from antibody-mediated injuries in an ex vivo simulation of ABO-incompatible kidney transplantation. Encouraged by these results, a discarded type B kidney, following enzymatic conversion, is transplanted into a type O brain-dead recipient with high titer of anti-B antibody. The allograft survives for 63 hours without hyperacute rejection. Blood group B antigens re-express within 48 hours, with histopathological analyses indicating no evidence of antibody-mediated rejection. This enzymatic conversion approach holds the potential to broaden the practice of ABO-incompatible kidney transplantation, decrease waiting times and facilitate equitable organ allocation.
Collapse
Affiliation(s)
- Jun Zeng
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ming Ma
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Transplant Center and NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojuan Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhengsheng Rao
- Center of urology and nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Huang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Saifu Yin
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Bao
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haohan Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiling Wang
- Center of urology and nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongwei Gao
- Department of Blood Biochemistry and Molecular Biology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Feng Gong
- Department of Blood Biochemistry and Molecular Biology, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Tao Lin
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Keqin Zhang
- Center of urology and nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Turun Song
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Kidney Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
Zhou M, Luo K, Su C, Sun Y, Huang Z, Ma S, Gao X, Wang J, Zhang C, Han P, Wu G. Structural basis of FpGalNase and its combination with FpGalNAcDeAc for efficient A-to-O blood group conversion. Exp Hematol Oncol 2025; 14:7. [PMID: 39856738 PMCID: PMC11762096 DOI: 10.1186/s40164-025-00599-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Transfusion safety and blood typing continue to present significant challenges in clinical practice, including risks of incorrect blood transfusions and blood shortages. One promising solution is the enzymatic conversion of all red blood cell (RBC) types into universal O-type RBCs. However, the major obstacle to this strategy is the relatively low catalytic efficiency of the enzymes involved. In this study, we investigated two enzymes from Flavonifractor plautii, N-acetylgalactosamine deacetylase (FpGalNAcDeAc) and galactosaminidase (FpGalNase), which demonstrate synergistic activity in efficiently converting A-type RBCs to O-type. We optimized treatment conditions, achieving over 99% conversion in just five minutes using phosphate buffer saline and a 16 nM enzyme concentration. Additionally, we engineered two fusion proteins, FpGalNAcDeAc-FpGalNase and FpGalNase-FpGalNAcDeAc, which showed a 28-fold increase in catalytic efficiency compared to the enzyme mixture. Using cryo-electron microscopy, we resolved the full-length structure of FpGalNase, identifying critical active site residues involved in its catalytic mechanism. This study provides essential structural and biochemical insights for clinical applications in blood group conversion, offering a promising approach for producing universal O-type RBCs.
Collapse
Affiliation(s)
- Meiling Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Kaishan Luo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Chao Su
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yan Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zuyan Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Shuo Ma
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xun Gao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jiwei Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Chen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Pengcheng Han
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| | - Guoqiu Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Advanced Institute for Life and Health, Center of Clinical Laboratory Medicine, Department of Pharmacy, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Herbold CW, Noell SE, Lee CK, Vickers CJ, Stott MB, Eisen JA, McDonald IR, Cary SC. Nutritional niches of potentially endemic, facultatively anaerobic heterotrophs from an isolated Antarctic terrestrial hydrothermal refugium elucidated through metagenomics. ENVIRONMENTAL MICROBIOME 2024; 19:104. [PMID: 39696719 DOI: 10.1186/s40793-024-00655-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Tramway Ridge, a geothermal Antarctic Specially Protected Area (elevation 3340 m) located near the summit of Mount Erebus, is home to a unique community composed of cosmopolitan surface-associated micro-organisms and abundant, poorly understood subsurface-associated microorganisms. Here, we use shotgun metagenomics to compare the functional capabilities of this community to those found elsewhere on Earth and to infer in situ diversity and metabolic capabilities of abundant subsurface taxa. RESULTS We found that the functional potential in this community is most similar to that found in terrestrial hydrothermal environments (hot springs, sediments) and that the two dominant organisms in the subsurface carry high rates of in situ diversity which was taken as evidence of potential endemicity. They were found to be facultative anaerobic heterotrophs that likely share a pool of nitrogenous organic compounds while specializing in different carbon compounds. CONCLUSIONS Metagenomic insights have provided a detailed understanding of the microbe-based ecosystem found in geothermally heated fumaroles at Tramway Ridge. This approach enabled us to compare Tramway Ridge with other microbial systems, identify potentially endemic taxa and elucidate the key metabolic pathways that may enable specific organisms to dominate the ecosystem.
Collapse
Affiliation(s)
- Craig W Herbold
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand
- International Centre for Terrestrial Antarctic Research, Te Whare Wānanga O Waikato - University of Waikato, Kirikiriroa - Hamilton, New Zealand
- Te Kura Pūtaiao Koiora - School of Biological Sciences, Te Whare Wānanga O Waitaha - University of Canterbury, Ōtautahi - Christchurch, New Zealand
| | - Stephen E Noell
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand
- International Centre for Terrestrial Antarctic Research, Te Whare Wānanga O Waikato - University of Waikato, Kirikiriroa - Hamilton, New Zealand
| | - Charles K Lee
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand
- International Centre for Terrestrial Antarctic Research, Te Whare Wānanga O Waikato - University of Waikato, Kirikiriroa - Hamilton, New Zealand
| | - Chelsea J Vickers
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand
| | - Matthew B Stott
- Te Kura Pūtaiao Koiora - School of Biological Sciences, Te Whare Wānanga O Waitaha - University of Canterbury, Ōtautahi - Christchurch, New Zealand
| | | | - Ian R McDonald
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand.
- International Centre for Terrestrial Antarctic Research, Te Whare Wānanga O Waikato - University of Waikato, Kirikiriroa - Hamilton, New Zealand.
| | - S Craig Cary
- Te Aka Mātuatua - School of Science, Te Whare Wānanga O Waikato - University of Waikato, Private Bag 3105, Kirikiriroa - Hamilton, 3240, New Zealand
- International Centre for Terrestrial Antarctic Research, Te Whare Wānanga O Waikato - University of Waikato, Kirikiriroa - Hamilton, New Zealand
| |
Collapse
|
7
|
Chakraborti S, Ghosh S. Bridging the gap: Exploring the microbial influence on forensic ABO typing discrepancies for enhanced investigative accuracy. Forensic Sci Int 2024; 365:112284. [PMID: 39514953 DOI: 10.1016/j.forsciint.2024.112284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Forensic serology has long been a cornerstone in forensic investigations, utilizing multidisciplinary approaches to identify and individualize biological evidence at crime scenes. However, the field faces challenges such as the potential for blood group changes in individuals affected by diseases or medical treatments, leading to false outcomes in prolonged legal cases. Additionally, the passive adsorption of bacterial products by red blood cells can result in blood group switching, emphasizing the need for careful analysis, especially in cases of concurrent infections. Furthermore, post-mortem samples may exhibit blood group variations, necessitating meticulous examination and interpretation of forensic serology results. Despite the advent of DNA typing techniques, traditional forensic serology methods remain relevant, with skills in recognizing stain patterns and selecting informative specimens indispensable. The present review highlights the increasing recognition of bacterial contaminations and their implications for forensic serology, underscoring the need for ongoing research, innovation, and collaboration within the field. This understanding revitalizes forensic science by emphasizing the critical role of maintaining the integrity of serological analyses and enhancing overall reliability.
Collapse
Affiliation(s)
- Sreemoyee Chakraborti
- Department of Forensic Science Laboratory, Biology Division, Government of West Bengal, 37/1/2 Belgachia Road, Kolkata 700037, India.
| | - Sandip Ghosh
- Department of Forensic Science Laboratory, Biology Division, Government of West Bengal, 37/1/2 Belgachia Road, Kolkata 700037, India
| |
Collapse
|
8
|
Nou NO, Covington JK, Lai D, Mayali X, Seymour CO, Johnston J, Jiao JY, Buessecker S, Mosier D, Muok AR, Torosian N, Cook AM, Briegel A, Woyke T, Eloe-Fadrosh E, Shapiro N, Bryan SG, Sleezer S, Dimapilis J, Gonzalez C, Gonzalez L, Noriega M, Hess M, Carlson RP, Liu L, Li MM, Lian ZH, Zhu S, Liu F, Sun X, Gao B, Mewalal R, Harmon-Smith M, Blaby IK, Cheng JF, Weber PK, Grigorean G, Li WJ, Dekas AE, Pett-Ridge J, Dodsworth JA, Palmer M, Hedlund BP. Genome-guided isolation of the hyperthermophilic aerobe Fervidibacter sacchari reveals conserved polysaccharide metabolism in the Armatimonadota. Nat Commun 2024; 15:9534. [PMID: 39496591 PMCID: PMC11535203 DOI: 10.1038/s41467-024-53784-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
Few aerobic hyperthermophilic microorganisms degrade polysaccharides. Here, we describe the genome-enabled enrichment and optical tweezer-based isolation of an aerobic polysaccharide-degrading hyperthermophile, Fervidibacter sacchari, previously ascribed to candidate phylum Fervidibacteria. F. sacchari uses polysaccharides and monosaccharides for growth at 65-87.5 °C and expresses 191 carbohydrate-active enzymes (CAZymes) according to RNA-Seq and proteomics, including 31 with unusual glycoside hydrolase domains (GH109, GH177, GH179). Fluorescence in-situ hybridization and nanoscale secondary ion mass spectrometry confirmed rapid assimilation of 13C-starch in spring sediments. Purified GHs were optimally active at 80-100 °C on ten different polysaccharides. Finally, we propose reassigning Fervidibacteria as a class within phylum Armatimonadota, along with 18 other species, and show that a high number and diversity of CAZymes is a hallmark of the phylum, in both aerobic and anaerobic lineages. Our study establishes Fervidibacteria as hyperthermophilic polysaccharide degraders in terrestrial geothermal springs and suggests a broad role for Armatimonadota in polysaccharide catabolism.
Collapse
Affiliation(s)
- Nancy O Nou
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | | | - Dengxun Lai
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Xavier Mayali
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Cale O Seymour
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Juliet Johnston
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Jian-Yu Jiao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory, Sun Yat-Sen University, Zhuhai, PR China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Steffen Buessecker
- Department of Earth System Science, Stanford University, Stanford, CA, USA
| | - Damon Mosier
- Department of Biology, California State University, San Bernardino, CA, USA
- Department of Earth, Energy, and Environment, University of Calgary, Calgary, AB, Canada
| | - Alise R Muok
- Department of Microbial Sciences, Institute of Biology, Leiden University, Sylviusweg 72, Leiden, The Netherlands
| | - Nicole Torosian
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Allison M Cook
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Ariane Briegel
- Department of Microbial Sciences, Institute of Biology, Leiden University, Sylviusweg 72, Leiden, The Netherlands
| | - Tanja Woyke
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- University of California Merced, Life and Environmental Sciences, Merced, CA, USA
| | - Emiley Eloe-Fadrosh
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Nicole Shapiro
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Scott G Bryan
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Savannah Sleezer
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Joshua Dimapilis
- Department of Biology, California State University, San Bernardino, CA, USA
| | - Cristina Gonzalez
- Department of Biology, California State University, San Bernardino, CA, USA
| | - Lizett Gonzalez
- Department of Biology, California State University, San Bernardino, CA, USA
| | - Marlene Noriega
- Department of Biology, California State University, San Bernardino, CA, USA
| | - Matthias Hess
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - Ross P Carlson
- Chemical and Biological Engineering, Montana State University, Bozeman, MT, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Lan Liu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory, Sun Yat-Sen University, Zhuhai, PR China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Meng-Meng Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory, Sun Yat-Sen University, Zhuhai, PR China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Zheng-Han Lian
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory, Sun Yat-Sen University, Zhuhai, PR China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Siqi Zhu
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Provincial Observation and Research Station for Coastal Upwelling Ecosystem, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
| | - Fan Liu
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Provincial Observation and Research Station for Coastal Upwelling Ecosystem, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Xian Sun
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Provincial Observation and Research Station for Coastal Upwelling Ecosystem, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
| | - Beile Gao
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
- Guangdong Provincial Observation and Research Station for Coastal Upwelling Ecosystem, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, PR China
| | - Ritesh Mewalal
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Miranda Harmon-Smith
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Ian K Blaby
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Jan-Fang Cheng
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Peter K Weber
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Wen-Jun Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory, Sun Yat-Sen University, Zhuhai, PR China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
- State Key Laboratory of Desert and Oasis Ecology, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi, PR China
| | - Anne E Dekas
- Department of Earth System Science, Stanford University, Stanford, CA, USA
| | - Jennifer Pett-Ridge
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Jeremy A Dodsworth
- Department of Biology, California State University, San Bernardino, CA, USA
| | - Marike Palmer
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA.
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada.
| | - Brian P Hedlund
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, USA.
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
9
|
Lei C, Li Z, Ma S, Zhang Q, Guo J, Ouyang Q, Lei Q, Zhou L, Yang J, Lin J, Ettlinger R, Wuttke S, Li X, Brinker CJ, Zhu W. Improving normothermic machine perfusion and blood transfusion through biocompatible blood silicification. Proc Natl Acad Sci U S A 2024; 121:e2322418121. [PMID: 39159377 PMCID: PMC11363281 DOI: 10.1073/pnas.2322418121] [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: 12/22/2023] [Accepted: 07/11/2024] [Indexed: 08/21/2024] Open
Abstract
The growing world population and increasing life expectancy are driving the need to improve the quality of blood transfusion, organ transplantation, and preservation. Here, to improve the ability of red blood cells (RBCs) for normothermic machine perfusion, a biocompatible blood silicification approach termed "shielding-augmenting RBC-in-nanoscale amorphous silica (SARNAS)" has been developed. The key to RBC surface engineering and structure augmentation is the precise control of the hydrolysis form of silicic acid to realize stabilization of RBC within conformal nanoscale silica-based exoskeletons. The formed silicified RBCs (Si-RBCs) maintain membrane/structural integrity, normal cellular functions (e.g., metabolism, oxygen-carrying capability), and enhance resistance to external stressors as well as tunable mechanical properties, resulting in nearly 100% RBC cryoprotection. In vivo experiments confirm their excellent biocompatibility. By shielding RBC surface antigens, the Si-RBCs provide universal blood compatibility, the ability for allogeneic mechanical perfusion, and more importantly, the possibility for cross-species transfusion. Being simple, reliable, and easily scalable, the SARNAS strategy holds great promise to revolutionize the use of engineered blood for future clinical applications.
Collapse
Affiliation(s)
- Chuanyi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, People’s Republic of China
| | - Zeyu Li
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, People’s Republic of China
| | - Shuhao Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Department of Engineering Mechanics, and Center for X-Mechanics, Zhejiang University, Hangzhou310027, People’s Republic of China
| | - Qi Zhang
- The Second Affiliated Hospital of Anhui Medical University, Hefei23060, People’s Republic of China
| | - Jimin Guo
- Center for Micro-Engineered Materials and the Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, NM87131
| | - Qing Ouyang
- Department of Hepatobiliary Surgery and Liver Transplant Center, The General Hospital of Southern Theater, Guangzhou510010, People’s Republic of China
| | - Qi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, People’s Republic of China
| | - Liang Zhou
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, People’s Republic of China
| | - Junxian Yang
- Research Department of Medical Sciences, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou510080, China
| | - Jiangguo Lin
- Research Department of Medical Sciences, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou510080, China
| | - Romy Ettlinger
- EastChem School of Chemistry, University of St Andrews, North Haugh, St. AndrewsKY16 9ST, United Kingdom
| | - Stefan Wuttke
- BCMaterials, Basque Center for Materials, UPV/EHU Science Park, Leioa48950, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao48009, Spain
| | - Xuejin Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, Department of Engineering Mechanics, and Center for X-Mechanics, Zhejiang University, Hangzhou310027, People’s Republic of China
| | - C. Jeffrey Brinker
- Center for Micro-Engineered Materials and the Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, NM87131
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou510006, People’s Republic of China
| |
Collapse
|
10
|
Kashima T, Akama M, Wakinaka T, Arakawa T, Ashida H, Fushinobu S. Crystal Structure of Bifidobacterium bifidum Glycoside Hydrolase Family 110 α-Galactosidase Specific for Blood Group B Antigen. J Appl Glycosci (1999) 2024; 71:81-90. [PMID: 39234034 PMCID: PMC11368712 DOI: 10.5458/jag.jag.jag-2024_0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/18/2024] [Indexed: 09/06/2024] Open
Abstract
To overcome incompatibility issues and increase the possibility of blood transfusion, technologies that enable efficient conversion of A- and B-type red blood cells to the universal donor O-type is desirable. Although several blood type-converting enzymes have been identified, detailed understanding about their molecular functions is limited. α-Galactosidase from Bifidobacterium bifidum JCM 1254 (AgaBb), belonging to glycoside hydrolase (GH) 110 subfamily A, specifically acts on blood group B antigen. Here we present the crystal structure of AgaBb, including the catalytic GH110 domain and part of the C-terminal uncharacterized regions. Based on this structure, we deduced a possible binding mechanism of blood group B antigen to the active site. Site-directed mutagenesis confirmed that R270 and E380 recognize the fucose moiety in the B antigen. Thermal shift assay revealed that the C-terminal uncharacterized region significantly contributes to protein stability. This region is shared only among GH110 enzymes from B. bifidum and some Ruminococcus species. The elucidation of the molecular basis for the specific recognition of blood group B antigen is expected to lead to the practical application of blood group conversion enzymes in the future.
Collapse
Affiliation(s)
- Toma Kashima
- Department of Biotechnology, The University of Tokyo
| | - Megumi Akama
- Department of Biotechnology, The University of Tokyo
| | | | | | - Hisashi Ashida
- Faculty of Biology-Oriented Science and Technology, Kindai University
| | - Shinya Fushinobu
- Department of Biotechnology, The University of Tokyo
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo
| |
Collapse
|
11
|
Jiang J, Czuchry D, Ru Y, Peng H, Shen J, Wang T, Zhao W, Chen W, Sui SF, Li Y, Li N. Activity-based metaproteomics driven discovery and enzymological characterization of potential α-galactosidases in the mouse gut microbiome. Commun Chem 2024; 7:184. [PMID: 39152233 PMCID: PMC11329505 DOI: 10.1038/s42004-024-01273-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
The gut microbiota offers an extensive resource of enzymes, but many remain uncharacterized. To distinguish the activities of similar annotated proteins and mine the potentially applicable ones in the microbiome, we applied an effective Activity-Based Metaproteomics (ABMP) strategy using a specific activity-based probe (ABP) to screen the entire gut microbiome for directly discovering active enzymes and their potential applications, not for exploring host-microbiome interactions. By using an activity-based cyclophellitol aziridine probe specific to α-galactosidases (AGAL), we successfully identified and characterized several gut microbiota enzymes possessing AGAL activities. Cryo-electron microscopy analysis of a newly characterized enzyme (AGLA5) revealed the covalent binding conformations between the AGAL5 active site and the cyclophellitol aziridine ABP, which could provide insights into the enzyme's catalytic mechanism. The four newly characterized AGALs have diverse potential activities, including raffinose family oligosaccharides (RFOs) hydrolysis and enzymatic blood group transformation. Collectively, we present a ABMP platform that facilitates gut microbiota AGALs discovery, biochemical activity annotations and potential industrial or biopharmaceutical applications.
Collapse
Affiliation(s)
- Jianbing Jiang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Diana Czuchry
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yanxia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Huipai Peng
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junfeng Shen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Teng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wenjuan Zhao
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Weihua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Sen-Fang Sui
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yaowang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Nan Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Shenzhen, China.
| |
Collapse
|
12
|
Yang J, Li A, Li M, Ruan S, Ye L. CRISPR/Cas9-Editing K562 Cell Line as a Potential Tool in Transfusion Applications: Knockout of Vel Antigen Gene. Transfus Med Hemother 2024; 51:265-273. [PMID: 39021420 PMCID: PMC11250041 DOI: 10.1159/000534012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/04/2023] [Indexed: 07/20/2024] Open
Abstract
Introduction The Vel- phenotype is a rare blood group, and it is challenging for identifying this phenotype due to limited available reagents. Moreover, there are relatively few studies on genomic editing of erythroid antigens and generation of knockout (KO) cell lines at present. Methods To identify the high-efficiency small-guiding RNA (sgRNA) sequence, candidate sgRNAs were transfected into HEK 293T cells and analyzed using Sanger sequencing. Following this, the high-efficiency sgRNA was transfected into K562 cells using lentivirus transduction to generate KO Vel blood group gene cells. The expression of the Vel protein was detected using Western blot on single-cell clones. Additionally, flow cytometry was used to detect the erythroid markers CD235a and CD71. Hemoglobin quantification and Giemsa staining were also performed to evaluate the erythroid differentiation of KO clones induced by hemin. Results The high-efficiency sgRNA was successfully obtained and used for CRISPR-Cas9 editing in K562 cells. After limiting dilution and screening, two KO clones had either deleted 2 or 4 bases and showed no expression of the Vel protein. In the hemin-induced KO clone, there was a significant difference in erythroid marker and hemoglobin quantification compared to untreated cells. The morphological changes were also observed for the hemin-induced KO clone. Conclusion In this study, a highly efficient sgRNA was screened out and used to generate Vel erythroid antigen KO single-cell clones in K562 cells. The edited cells could then be induced to undergo erythroid differentiation with the use of hemin.
Collapse
Affiliation(s)
- Jiaxuan Yang
- Molecular Immunohematology Lab, Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai, China
| | - Aijing Li
- Molecular Immunohematology Lab, Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai, China
| | - Minghao Li
- Molecular Immunohematology Lab, Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai, China
| | - Shulin Ruan
- Molecular Immunohematology Lab, Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai, China
| | - Luyi Ye
- Molecular Immunohematology Lab, Shanghai Institute of Blood Transfusion, Shanghai Blood Center, Shanghai, China
| |
Collapse
|
13
|
Mudhluli TE, Kujawska M, Mueller J, Felsl A, Truppel BA, Hall LJ, Chitsike I, Gomo E, Zhou DT. Exploring the genomic traits of infant-associated microbiota members from a Zimbabwean cohort. BMC Genomics 2024; 25:718. [PMID: 39054474 PMCID: PMC11271062 DOI: 10.1186/s12864-024-10618-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
INTRODUCTION Our understanding of particular gut microbiota members such as Bifidobacterium and Enterococcus in low-middle-income countries remains very limited, particularly early life strain-level beneficial traits. This study addresses this gap by exploring a collection of bacterial strains isolated from the gut of Zimbabwean infants; comparing their genomic characteristics with strains isolated from infants across North America, Europe, and other regions of Africa. MATERIALS AND METHOD From 110 infant stool samples collected in Harare, Zimbabwe, 20 randomly selected samples were used to isolate dominant early-life gut microbiota members Bifidobacterium and Enterococcus. Isolated strains were subjected to whole genome sequencing and bioinformatics analysis including functional annotation of carbohydrates, human milk oligosaccharide (HMO) and protein degradation genes and clusters, and the presence of antibiotic resistance genes (ARGs). RESULTS The study observed some location-based clustering within the main five identified taxonomic groups. Furthermore, there were varying and overall species-specific numbers of genes belonging to different GH families encoded within the analysed dataset. Additionally, distinct strain- and species-specific variances were identified in the potential of Bifidobacterium for metabolizing HMOs. Analysis of putative protease activity indicated a consistent presence of gamma-glutamyl hydrolases in Bifidobacterium, while Enterococcus genomes exhibited a high abundance of aspartyl peptidases. Both genera harboured resistance genes against multiple classes of antimicrobial drugs, with Enterococcus genomes containing a higher number of ARGs compared to Bifidobacterium, on average. CONCLUSION This study identified promising probiotic strains within Zimbabwean isolates, offering the potential for early-life diet and microbial therapies. However, the presence of antibiotic resistance genes in infant-associated microbes raises concerns for infection risk and next-stage probiotic development. Further investigation in larger cohorts, particularly in regions with limited existing data on antibiotic and probiotic use, is crucial to validate these initial insights. IMPACT STATEMENT This research represents the first investigation of its kind in the Zimbabwean context, focusing on potential probiotic strains within the early-life gut microbiota. By identifying local probiotic strains, this research can contribute to the development of probiotic interventions that are tailored to the Zimbabwean population, which can help address local health challenges and promote better health outcomes for infants. Another essential aspect of the study is the investigation of antimicrobial resistance genes present in Zimbabwean bacterial strains. Antimicrobial resistance is a significant global health concern, and understanding the prevalence and distribution of resistance genes in different regions can help inform public health policies and interventions.
Collapse
Affiliation(s)
- Taona Emmah Mudhluli
- Faculty of Medicine and Health Sciences, Department of Laboratory Diagnostic and Investigative Sciences, Medical Laboratory Sciences Unit, University of Zimbabwe, Box A 178, Avondale, Harare, Zimbabwe.
- Faculty of Medicine and Health Science, Department of Biochemistry, Midlands State University, P. Bag 9055, Senga Road, Gweru, Zimbabwe.
| | - Magdalena Kujawska
- Intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Julia Mueller
- Intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Angela Felsl
- Intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Bastian-Alexander Truppel
- Intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
- BioSciences Building, APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
| | - Lindsay J Hall
- Intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
- Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, B15 42TT, UK
| | - Inam Chitsike
- Faculty of Medicine and Health Sciences, Department of Family Health, Paediatrics Unit, University of Zimbabwe, Box A 178, Avondale, Harare, Zimbabwe
| | - Exnevia Gomo
- Faculty of Medicine and Health Sciences, Department of Laboratory Diagnostic and Investigative Sciences, Medical Laboratory Sciences Unit, University of Zimbabwe, Box A 178, Avondale, Harare, Zimbabwe
| | - Danai Tavonga Zhou
- Faculty of Medicine and Health Sciences, Department of Laboratory Diagnostic and Investigative Sciences, Medical Laboratory Sciences Unit, University of Zimbabwe, Box A 178, Avondale, Harare, Zimbabwe
| |
Collapse
|
14
|
Wardman JF, Withers SG. Carbohydrate-active enzyme (CAZyme) discovery and engineering via (Ultra)high-throughput screening. RSC Chem Biol 2024; 5:595-616. [PMID: 38966674 PMCID: PMC11221537 DOI: 10.1039/d4cb00024b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
Carbohydrate-active enzymes (CAZymes) constitute a diverse set of enzymes that catalyze the assembly, degradation, and modification of carbohydrates. These enzymes have been fashioned into potent, selective catalysts by millennia of evolution, and yet are also highly adaptable and readily evolved in the laboratory. To identify and engineer CAZymes for different purposes, (ultra)high-throughput screening campaigns have been frequently utilized with great success. This review provides an overview of the different approaches taken in screening for CAZymes and how mechanistic understandings of CAZymes can enable new approaches to screening. Within, we also cover how cutting-edge techniques such as microfluidics, advances in computational approaches and synthetic biology, as well as novel assay designs are leading the field towards more informative and effective screening approaches.
Collapse
Affiliation(s)
- Jacob F Wardman
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver BC V6T 1Z3 Canada
- Michael Smith Laboratories, University of British Columbia Vancouver BC V6T 1Z4 Canada
| | - Stephen G Withers
- Department of Biochemistry and Molecular Biology, University of British Columbia Vancouver BC V6T 1Z3 Canada
- Michael Smith Laboratories, University of British Columbia Vancouver BC V6T 1Z4 Canada
- Department of Chemistry, University of British Columbia Vancouver BC V6T 1Z1 Canada
| |
Collapse
|
15
|
Nasseri SA, Lazarski AC, Lemmer IL, Zhang CY, Brencher E, Chen HM, Sim L, Panwar D, Betschart L, Worrall LJ, Brumer H, Strynadka NCJ, Withers SG. An alternative broad-specificity pathway for glycan breakdown in bacteria. Nature 2024; 631:199-206. [PMID: 38898276 DOI: 10.1038/s41586-024-07574-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
The vast majority of glycosidases characterized to date follow one of the variations of the 'Koshland' mechanisms1 to hydrolyse glycosidic bonds through substitution reactions. Here we describe a large-scale screen of a human gut microbiome metagenomic library using an assay that selectively identifies non-Koshland glycosidase activities2. Using this, we identify a cluster of enzymes with extremely broad substrate specificities and thoroughly characterize these, mechanistically and structurally. These enzymes not only break glycosidic linkages of both α and β stereochemistry and multiple connectivities, but also cleave substrates that are not hydrolysed by standard glycosidases. These include thioglycosides, such as the glucosinolates from plants, and pseudoglycosidic bonds of pharmaceuticals such as acarbose. This is achieved through a distinct mechanism of hydrolysis that involves oxidation/reduction and elimination/hydration steps, each catalysed by enzyme modules that are in many cases interchangeable between organisms and substrate classes. Homologues of these enzymes occur in both Gram-positive and Gram-negative bacteria associated with the gut microbiome and other body parts, as well as other environments, such as soil and sea. Such alternative step-wise mechanisms appear to constitute largely unrecognized but abundant pathways for glycan degradation as part of the metabolism of carbohydrates in bacteria.
Collapse
Affiliation(s)
- Seyed Amirhossein Nasseri
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aleksander C Lazarski
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Center for Blood Research, University of British Columbia, Vancouver, Canada
| | - Imke L Lemmer
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chloe Y Zhang
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eva Brencher
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hong-Ming Chen
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lyann Sim
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Deepesh Panwar
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leo Betschart
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liam J Worrall
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Center for Blood Research, University of British Columbia, Vancouver, Canada
| | - Harry Brumer
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Center for Blood Research, University of British Columbia, Vancouver, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
16
|
Kim H, Mi HTN, Ahn JH, Lee JS, Eser BE, Choi J, Han J. Glycoside-metabolizing oxidoreductase D3dgpA from human gut bacterium. Front Bioeng Biotechnol 2024; 12:1413854. [PMID: 39007053 PMCID: PMC11239390 DOI: 10.3389/fbioe.2024.1413854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/20/2024] [Indexed: 07/16/2024] Open
Abstract
The Gfo/Idh/MocA family enzyme DgpA was known to catalyze the regiospecific oxidation of puerarin to 3"-oxo-puerarin in the presence of 3-oxo-glucose. Here, we discovered that D3dgpA, dgpA cloned from the human gut bacterium Dorea sp. MRG-IFC3, catalyzed the regiospecific oxidation of various C-/O-glycosides, including puerarin, in the presence of methyl β-D-3-oxo-glucopyranoside. While C-glycosides were converted to 3"- and 2"-oxo-products by D3dgpA, O-glycosides resulted in the formation of aglycones and hexose enediolone from the 3"-oxo-products. From DFT calculations, it was found that isomerization of 3"-oxo-puerarin to 2"-oxo-puerarin required a small activation energy of 9.86 kcal/mol, and the O-glycosidic bond cleavage of 3"-oxo-products was also thermodynamically favored with a small activation energy of 3.49 kcal/mol. In addition, the reaction mechanism of D3dgpA was discussed in comparison to those of Gfo/Idh/MocA and GMC family enzymes. The robust reactivity of D3dgpA was proposed as a new general route for derivatization of glycosides.
Collapse
Affiliation(s)
- Heji Kim
- Metalloenzyme Research Group and Department of Plant Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Huynh Thi Ngoc Mi
- Metalloenzyme Research Group and Department of Plant Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Joong-Hoon Ahn
- Department of Integrative Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Jong Suk Lee
- Bio Industry Department, Gyeonggido Business and Science Accelerator (GBSA), Suwon, Gyeonngi-do, Republic of Korea
| | - Bekir Engin Eser
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | - Jongkeun Choi
- Department of Chemical Engineering, Chungwoon University, Incheon, Republic of Korea
| | - Jaehong Han
- Metalloenzyme Research Group and Department of Plant Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| |
Collapse
|
17
|
Bains RK, Nasseri SA, Wardman JF, Withers SG. Advances in the understanding and exploitation of carbohydrate-active enzymes. Curr Opin Chem Biol 2024; 80:102457. [PMID: 38657391 DOI: 10.1016/j.cbpa.2024.102457] [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: 02/06/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
Carbohydrate-active enzymes (CAZymes) are responsible for the biosynthesis, modification and degradation of all glycans in Nature. Advances in genomic and metagenomic methodologies, in conjunction with lower cost gene synthesis, have provided access to a steady stream of new CAZymes with both well-established and novel mechanisms. At the same time, increasing access to cryo-EM has resulted in exciting new structures, particularly of transmembrane glycosyltransferases of various sorts. This improved understanding has resulted in widespread progress in applications of CAZymes across diverse fields, including therapeutics, organ transplantation, foods, and biofuels. Herein, we highlight a few of the many important advances that have recently been made in the understanding and applications of CAZymes.
Collapse
Affiliation(s)
- Rajneesh K Bains
- Department of Chemistry, University of British Columbia, Vancouver, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Seyed Amirhossein Nasseri
- Department of Chemistry, University of British Columbia, Vancouver, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Jacob F Wardman
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada; Department of Biochemistry, University of British Columbia, Vancouver, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, Canada; Department of Biochemistry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
18
|
Wang R, Li X, Lv F, He J, Lv R, Wei L. Sesame bacterial wilt significantly alters rhizosphere soil bacterial community structure, function, and metabolites in continuous cropping systems. Microbiol Res 2024; 282:127649. [PMID: 38402727 DOI: 10.1016/j.micres.2024.127649] [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: 01/01/2024] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Bacterial wilt is the leading disease of sesame and alters the bacterial community composition, function, and metabolism of sesame rhizosphere soil. However, its pattern of change is unclear. Here, the purpose of this study was to investigate how these communities respond to three differing severities of bacterial wilt in mature continuously cropped sesame plants by metagenomic and metabolomic techniques, namely, absence (WH), moderate (WD5), and severe (WD9) wilt. The results indicated that bacterial wilt could significantly change the bacterial community structure in the rhizosphere soil of continuously cropped sesame plants. The biomarker species with significant differences will also change with increasing disease severity. In particular, the gene expression levels of Ralstonia solanacearum in the WD9 and WD5 treatments increased by 25.29% and 33.61%, respectively, compared to those in the WH treatment (4.35 log10 copies g-1). The occurrence of bacterial wilt significantly altered the functions of the bacterial community in rhizosphere soil. KEEG and CAZy functional annotations revealed that the number of significantly different functions in WH was greater than that in WD5 and WD9. Bacterial wilt significantly affected the relative content of metabolites, especially acids, in the rhizosphere soil, and compared with those in the rhizosphere soil from WH, 10 acids (including S-adenosylmethionine, N-acetylleucine, and desaminotyrosine, etc.) in the rhizosphere soil from WD5 or WD9 significantly increased. In comparison, the changes in the other 10 acids (including hypotaurine, erucic acid, and 6-hydroxynicotinic acid, etc.) were reversed. The occurrence of bacterial wilt also significantly inhibited metabolic pathways such as ABC transporter and amino acid biosynthesis pathways in rhizosphere soil and had a significant impact on two key enzymes (1.1.1.11 and 2.6.1.44). In conclusion, sesame bacterial wilt significantly alters the rhizosphere soil bacterial community structure, function, and metabolites. This study enhances the understanding of sesame bacterial wilt mechanisms and lays the groundwork for future prevention and control strategies against this disease.
Collapse
Affiliation(s)
- Ruiqing Wang
- Soil Fertilizer and Resource Environment Institute, Jiangxi Academy of Agricultural Sciences, No. 602, Nanlian Road, Nanchang, Jiangxi Province 330200, PR China; Key Laboratory of Crop Ecophysiology and Farming System for the Middle and Lower Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, PR China; National Engineering Technology Research Center for Red Soil Improvement, PR China; National Agricultural Experimental Station for Agricultural Environment Yichun, PR China.
| | - Xinsheng Li
- Institute of Plant Protection, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi Province 330200, PR China
| | - Fengjuan Lv
- Soil Fertilizer and Resource Environment Institute, Jiangxi Academy of Agricultural Sciences, No. 602, Nanlian Road, Nanchang, Jiangxi Province 330200, PR China; Key Laboratory of Crop Ecophysiology and Farming System for the Middle and Lower Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, PR China; National Engineering Technology Research Center for Red Soil Improvement, PR China; National Agricultural Experimental Station for Agricultural Environment Yichun, PR China
| | - Junhai He
- Soil Fertilizer and Resource Environment Institute, Jiangxi Academy of Agricultural Sciences, No. 602, Nanlian Road, Nanchang, Jiangxi Province 330200, PR China; Key Laboratory of Crop Ecophysiology and Farming System for the Middle and Lower Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, PR China; National Engineering Technology Research Center for Red Soil Improvement, PR China; National Agricultural Experimental Station for Agricultural Environment Yichun, PR China
| | - Rujie Lv
- Soil Fertilizer and Resource Environment Institute, Jiangxi Academy of Agricultural Sciences, No. 602, Nanlian Road, Nanchang, Jiangxi Province 330200, PR China; Key Laboratory of Crop Ecophysiology and Farming System for the Middle and Lower Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, PR China; National Engineering Technology Research Center for Red Soil Improvement, PR China; National Agricultural Experimental Station for Agricultural Environment Yichun, PR China
| | - Lingen Wei
- Soil Fertilizer and Resource Environment Institute, Jiangxi Academy of Agricultural Sciences, No. 602, Nanlian Road, Nanchang, Jiangxi Province 330200, PR China; Key Laboratory of Crop Ecophysiology and Farming System for the Middle and Lower Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, PR China; National Engineering Technology Research Center for Red Soil Improvement, PR China; National Agricultural Experimental Station for Agricultural Environment Yichun, PR China.
| |
Collapse
|
19
|
Crouch LI, Rodrigues CS, Bakshani CR, Tavares-Gomes L, Gaifem J, Pinho SS. The role of glycans in health and disease: Regulators of the interaction between gut microbiota and host immune system. Semin Immunol 2024; 73:101891. [PMID: 39388764 DOI: 10.1016/j.smim.2024.101891] [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: 08/02/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
The human gut microbiota is home to a diverse collection of microorganisms that has co-evolved with the host immune system in which host-microbiota interactions are essential to preserve health and homeostasis. Evidence suggests that the perturbation of this symbiotic host-microbiome relationship contributes to the onset of major diseases such as chronic inflammatory diseases including Inflammatory Bowel Disease. The host glycocalyx (repertoire of glycans/sugar-chains at the surface of gut mucosa) constitutes a major biological and physical interface between the intestinal mucosa and microorganisms, as well as with the host immune system. Glycans are an essential niche for microbiota colonization and thus an important modulator of host-microorganism interactions both in homeostasis and in disease. In this review, we discuss the role of gut mucosa glycome as an instrumental pathway that regulates host-microbiome interactions in homeostasis but also in health to inflammation transition. We also discuss the power of mucosa glycosylation remodelling as an attractive preventive and therapeutic strategy to preserve gut homeostasis.
Collapse
Affiliation(s)
- Lucy I Crouch
- Department of Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK.
| | - Cláudia S Rodrigues
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Cassie R Bakshani
- Department of Microbes, Infection and Microbiomes, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Leticia Tavares-Gomes
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
20
|
Jensen M, Stenfelt L, Ricci Hagman J, Pichler MJ, Weikum J, Nielsen TS, Hult A, Morth JP, Olsson ML, Abou Hachem M. Akkermansia muciniphila exoglycosidases target extended blood group antigens to generate ABO-universal blood. Nat Microbiol 2024; 9:1176-1188. [PMID: 38684911 DOI: 10.1038/s41564-024-01663-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/04/2024] [Indexed: 05/02/2024]
Abstract
Matching donor and recipient blood groups based on red blood cell (RBC) surface ABO glycans and antibodies in plasma is crucial to avoid potentially fatal reactions during transfusions. Enzymatic conversion of RBC glycans to the universal group O is an attractive solution to simplify blood logistics and prevent ABO-mismatched transfusions. The gut symbiont Akkermansia muciniphila can degrade mucin O-glycans including ABO epitopes. Here we biochemically evaluated 23 Akkermansia glycosyl hydrolases and identified exoglycosidase combinations which efficiently transformed both A and B antigens and four of their carbohydrate extensions. Enzymatic removal of canonical and extended ABO antigens on RBCs significantly improved compatibility with group O plasmas, compared to conversion of A or B antigens alone. Finally, structural analyses of two B-converting enzymes identified a previously unknown putative carbohydrate-binding module. This study demonstrates the potential utility of mucin-degrading gut bacteria as valuable sources of enzymes for production of universal blood for transfusions.
Collapse
Affiliation(s)
- Mathias Jensen
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Linn Stenfelt
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Jennifer Ricci Hagman
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Sweden
| | - Michael Jakob Pichler
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Julia Weikum
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Tine Sofie Nielsen
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Annika Hult
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Sweden
| | - Jens Preben Morth
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin L Olsson
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Department of Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Sweden.
| | - Maher Abou Hachem
- Department of Biotechnology & Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
21
|
Balumahendra K, Venkateswarulu TC, Babu DJ. Enhancement of α-galactosidase production using novel Actinoplanes utahensis B1 strain: sequential optimization and purification of enzyme. World J Microbiol Biotechnol 2024; 40:91. [PMID: 38345638 DOI: 10.1007/s11274-023-03880-1] [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: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 02/15/2024]
Abstract
α-Galactosidase is an important exoglycosidase belonging to the hydrolase class of enzymes, which has therapeutic and industrial potential. It plays a crucial role in hydrolyzing α-1,6 linked terminal galacto-oligosaccharide residues such as melibiose, raffinose, and branched polysaccharides such as galacto-glucomannans and galactomannans. In this study, Actinoplanes utahensis B1 was explored for α-galactosidase production, yield improvement, and activity enhancement by purification. Initially, nine media components were screened using the Plackett-Burman design (PBD). Among these components, sucrose, soya bean flour, and sodium glutamate were identified as the best-supporting nutrients for the highest enzyme secretion by A. Utahensis B1. Later, the Central Composite Design (CCD) was implemented to fine-tune the optimization of these components. Based on sequential statistical optimization methodologies, a significant, 3.64-fold increase in α-galactosidase production, from 16 to 58.37 U/mL was achieved. The enzyme was purified by ultrafiltration-I followed by multimode chromatography and ultrafiltration-II. The purity of the enzyme was confirmed by Sodium Dodecyl Sulphate-Polyacrylamide Agarose Gel Electrophoresis (SDS-PAGE) which revealed a single distinctive band with a molecular weight of approximately 72 kDa. Additionally, it was determined that this process resulted in a 2.03-fold increase in purity. The purified α-galactosidase showed an activity of 2304 U/mL with a specific activity of 288 U/mg. This study demonstrates the isolation of Actinoplanes utahensis B1 and optimization of the process for the α-galactosidase production as well as single-step purification.
Collapse
Affiliation(s)
- K Balumahendra
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur District, Guntur, Andhra Pradesh, India
| | - T C Venkateswarulu
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur District, Guntur, Andhra Pradesh, India
| | - D John Babu
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur District, Guntur, Andhra Pradesh, India.
| |
Collapse
|
22
|
Zhernakova DV, Wang D, Liu L, Andreu-Sánchez S, Zhang Y, Ruiz-Moreno AJ, Peng H, Plomp N, Del Castillo-Izquierdo Á, Gacesa R, Lopera-Maya EA, Temba GS, Kullaya VI, van Leeuwen SS, Xavier RJ, de Mast Q, Joosten LAB, Riksen NP, Rutten JHW, Netea MG, Sanna S, Wijmenga C, Weersma RK, Zhernakova A, Harmsen HJM, Fu J. Host genetic regulation of human gut microbial structural variation. Nature 2024; 625:813-821. [PMID: 38172637 PMCID: PMC10808065 DOI: 10.1038/s41586-023-06893-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 11/23/2023] [Indexed: 01/05/2024]
Abstract
Although the impact of host genetics on gut microbial diversity and the abundance of specific taxa is well established1-6, little is known about how host genetics regulates the genetic diversity of gut microorganisms. Here we conducted a meta-analysis of associations between human genetic variation and gut microbial structural variation in 9,015 individuals from four Dutch cohorts. Strikingly, the presence rate of a structural variation segment in Faecalibacterium prausnitzii that harbours an N-acetylgalactosamine (GalNAc) utilization gene cluster is higher in individuals who secrete the type A oligosaccharide antigen terminating in GalNAc, a feature that is jointly determined by human ABO and FUT2 genotypes, and we could replicate this association in a Tanzanian cohort. In vitro experiments demonstrated that GalNAc can be used as the sole carbohydrate source for F. prausnitzii strains that carry the GalNAc-metabolizing pathway. Further in silico and in vitro studies demonstrated that other ABO-associated species can also utilize GalNAc, particularly Collinsella aerofaciens. The GalNAc utilization genes are also associated with the host's cardiometabolic health, particularly in individuals with mucosal A-antigen. Together, the findings of our study demonstrate that genetic associations across the human genome and bacterial metagenome can provide functional insights into the reciprocal host-microbiome relationship.
Collapse
Affiliation(s)
- Daria V Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Daoming Wang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Lei Liu
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Yue Zhang
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Angel J Ruiz-Moreno
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Haoran Peng
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Niels Plomp
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Ángela Del Castillo-Izquierdo
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Esteban A Lopera-Maya
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Godfrey S Temba
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vesla I Kullaya
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Sander S van Leeuwen
- University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen, The Netherlands
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Serena Sanna
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- Institute for Genetic and Biomedical Research, National Research Council, Cagliari, Italy
| | - Cisca Wijmenga
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands.
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands.
| |
Collapse
|
23
|
Kaur A, Pickles IB, Sharma M, Madeido Soler N, Scott NE, Pidot SJ, Goddard-Borger ED, Davies GJ, Williams SJ. Widespread Family of NAD +-Dependent Sulfoquinovosidases at the Gateway to Sulfoquinovose Catabolism. J Am Chem Soc 2023; 145:28216-28223. [PMID: 38100472 PMCID: PMC10755693 DOI: 10.1021/jacs.3c11126] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
The sulfosugar sulfoquinovose (SQ) is produced by photosynthetic plants, algae, and cyanobacteria on a scale of 10 billion tons per annum. Its degradation, which is essential to allow cycling of its constituent carbon and sulfur, involves specialized glycosidases termed sulfoquinovosidases (SQases), which release SQ from sulfolipid glycoconjugates, so SQ can enter catabolism pathways. However, many SQ catabolic gene clusters lack a gene encoding a classical SQase. Here, we report the discovery of a new family of SQases that use an atypical oxidoreductive mechanism involving NAD+ as a catalytic cofactor. Three-dimensional X-ray structures of complexes with SQ and NAD+ provide insight into the catalytic mechanism, which involves transient oxidation at C3. Bioinformatic survey reveals this new family of NAD+-dependent SQases occurs within sulfoglycolytic and sulfolytic gene clusters that lack classical SQases and is distributed widely including within Roseobacter clade bacteria, suggesting an important contribution to marine sulfur cycling.
Collapse
Affiliation(s)
- Arashdeep Kaur
- School
of Chemistry, University of Melbourne, Parkville, Victoria 3010, Australia
- Bio21
Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Isabelle B. Pickles
- York
Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 5DD, U.K.
| | - Mahima Sharma
- York
Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 5DD, U.K.
| | - Niccolay Madeido Soler
- ACRF
Chemical Biology Division, The Walter and
Eliza Hall Institute of Medical Research, Parkville, Victoria 3010, Australia
- Department
of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nichollas E. Scott
- Department
of Microbiology and Immunology, University
of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Sacha J. Pidot
- Department
of Microbiology and Immunology, University
of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Ethan D. Goddard-Borger
- ACRF
Chemical Biology Division, The Walter and
Eliza Hall Institute of Medical Research, Parkville, Victoria 3010, Australia
- Department
of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Gideon J. Davies
- York
Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 5DD, U.K.
| | - Spencer J. Williams
- School
of Chemistry, University of Melbourne, Parkville, Victoria 3010, Australia
- Bio21
Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
24
|
Curci N, Iacono R, Segura DR, Cillo M, Cobucci-Ponzano B, Strazzulli A, Leonardi A, Giger L, Moracci M. Novel GH109 enzymes for bioconversion of group A red blood cells to the universal donor group O. N Biotechnol 2023; 77:130-138. [PMID: 37643666 DOI: 10.1016/j.nbt.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 08/31/2023]
Abstract
Glycoside hydrolases (GHs) have been employed for industrial and biotechnological purposes and often play an important role in new applications. The red blood cell (RBC) antigen system depends on the composition of oligosaccharides on the surface of erythrocytes, thus defining the ABO blood type classification. Incorrect blood transfusions may lead to fatal consequences, making the availability of the correct blood group critical. In this regard, it has been demonstrated that some GHs may be helpful in the conversion of groups A and B blood types to produce group O universal donor blood. GHs belonging to the GH109 family are of particular interest for this application due to their ability to convert blood from group A to group O. This work describes the biochemical characterisation of three novel GH109 enzymes (NAg68, NAg69 and NAg71) and the exploration of their ability to produce enzymatically converted RBCs (ECO-RBC). The three enzymes showed superior specificity on pNP-α-N-acetylgalactosamine compared to previously reported GH109 enzymes. These novel enzymes were able to act on purified antigen-A trisaccharides and produce ECO-RBC from human donor blood. NAg71 converted type A RBC to group O with increased efficiency in the presence of dextran compared to a commercially available GH109, previously used for this application.
Collapse
Affiliation(s)
- Nicola Curci
- Department of Biology, University of Naples "Federico II", Complesso Universitario di Monte S. Angelo, Via Cinthia 21, Naples 80126, Italy; Institute of Biosciences and BioResources, National Research Council of Italy, Via P. Castellino 111, Naples 80131, Italy
| | - Roberta Iacono
- Department of Biology, University of Naples "Federico II", Complesso Universitario di Monte S. Angelo, Via Cinthia 21, Naples 80126, Italy
| | | | - Michele Cillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Sergio Pansini, 5, Naples 80131, Italy
| | - Beatrice Cobucci-Ponzano
- Institute of Biosciences and BioResources, National Research Council of Italy, Via P. Castellino 111, Naples 80131, Italy
| | - Andrea Strazzulli
- Department of Biology, University of Naples "Federico II", Complesso Universitario di Monte S. Angelo, Via Cinthia 21, Naples 80126, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy
| | - Antonio Leonardi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Sergio Pansini, 5, Naples 80131, Italy
| | - Lars Giger
- Novozymes A/S, Biologiens vej 2, 2800 Kgs. Lyngby, Denmark
| | - Marco Moracci
- Department of Biology, University of Naples "Federico II", Complesso Universitario di Monte S. Angelo, Via Cinthia 21, Naples 80126, Italy; Institute of Biosciences and BioResources, National Research Council of Italy, Via P. Castellino 111, Naples 80131, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy.
| |
Collapse
|
25
|
Bitter J, Pfeiffer M, Borg AJE, Kuhlmann K, Pavkov-Keller T, Sánchez-Murcia PA, Nidetzky B. Enzymatic β-elimination in natural product O- and C-glycoside deglycosylation. Nat Commun 2023; 14:7123. [PMID: 37932298 PMCID: PMC10628242 DOI: 10.1038/s41467-023-42750-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
Biological degradation of natural product glycosides involves, alongside hydrolysis, β-elimination for glycosidic bond cleavage. Here, we discover an O-glycoside β-eliminase (OGE) from Agrobacterium tumefaciens that converts the C3-oxidized O-β-D-glucoside of phloretin (a plant-derived flavonoid) into the aglycone and the 2-hydroxy-3-keto-glycal elimination product. While unrelated in sequence, OGE is structurally homologous to, and shows effectively the same Mn2+ active site as, the C-glycoside deglycosylating enzyme (CGE) from a human intestinal bacterium implicated in β-elimination of 3-keto C-β-D-glucosides. We show that CGE catalyzes β-elimination of 3-keto O- and C-β-D-glucosides while OGE is specific for the O-glycoside substrate. Substrate comparisons and mutagenesis for CGE uncover positioning of aglycone for protonic assistance by the enzyme as critically important for C-glycoside cleavage. Collectively, our study suggests convergent evolution of active site for β-elimination of 3-keto O-β-D-glucosides. C-Glycoside cleavage is a specialized feature of this active site which is elicited by substrate through finely tuned enzyme-aglycone interactions.
Collapse
Affiliation(s)
- Johannes Bitter
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, A-8010, Graz, Austria
| | - Martin Pfeiffer
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, A-8010, Graz, Austria
| | - Annika J E Borg
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, A-8010, Graz, Austria
- Austrian Centre of Industrial Biotechnology, Krenngasse 37, A-8010, Graz, Austria
| | - Kirill Kuhlmann
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstraße 50/III, A-8010, Graz, Austria
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstraße 50/III, A-8010, Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, A-8010, Graz, Austria
- BioHealth Field of Excellence, University of Graz, Humboldtstraße 50, A-8010, Graz, Austria
| | - Pedro A Sánchez-Murcia
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingstalstraße 6/III, A-8010, Graz, Austria
| | - Bernd Nidetzky
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, A-8010, Graz, Austria.
- Austrian Centre of Industrial Biotechnology, Krenngasse 37, A-8010, Graz, Austria.
| |
Collapse
|
26
|
Ikegaya M, Park EY, Miyazaki T. Structure-function analysis of bacterial GH31 α-galactosidases specific for α-(1→4)-galactobiose. FEBS J 2023; 290:4984-4998. [PMID: 37438884 DOI: 10.1111/febs.16904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/21/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023]
Abstract
Glycoside hydrolase family 31 (GH31) contains α-glycoside hydrolases with different substrate specificities involved in various physiological functions. This family has recently been classified into 20 subfamilies using sequence similarity networks. An α-galactosidase from the gut bacterium Bacteroides salyersiae (BsGH31_19, which belongs to GH31 subfamily 19) was reported to have hydrolytic activity against the synthetic substrate p- nitrophenyl α-galactopyranoside, but its natural substrate remained unknown. BsGH31_19 shares low sequence identity (around 20%) with other reported GH31 α-galactosidases, PsGal31A from Pseudopedobacter saltans and human myogenesis-regulating glycosidase (MYORG), and was expected to have distinct specificity. Here, we characterized BsGH31_19 and its ortholog from a soil Bacteroidota bacterium, Flavihumibacter petaseus (FpGH31_19), and demonstrated that they showed high substrate specificity against α-(1→4)-linkages in α-(1→4)-galactobiose and globotriose [α-Gal-(1→4)-β-Gal-(1→4)-Glc], unlike PsGal31A and MYORG. The crystallographic analyses of BsGH31_19 and FpGH31_19 showed that their overall structures resemble those of MYORG and form a dimer with an interface different from that of PsGal31A and MYORG dimers. The structures of FpGH31_19 complexed with d-galactose and α-(1→4)-galactobiose revealed that amino acid residues that recognize a galactose residue at subsite +1 are not conserved between FpGH31_19 and BsGH31_19. The tryptophan (Trp153) that recognizes galactose at subsite -1 is homologous to the tryptophan residues in MYORG and α-galactosidases belonging to GH27, GH36, and GH97, but not in the bacterial GH31 member PsGal31A. Our results provide structural insights into molecular diversity and evolutionary relationships in the GH31 α-galactosidase subfamilies and the other α-galactosidase families.
Collapse
Affiliation(s)
- Marina Ikegaya
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, Japan
| | - Enoch Y Park
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, Japan
- Research Institute of Green Science and Technology, Shizuoka University, Japan
| | - Takatsugu Miyazaki
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, Japan
- Research Institute of Green Science and Technology, Shizuoka University, Japan
| |
Collapse
|
27
|
Choi J, Kim Y, Eser BE, Han J. Theoretical study on the glycosidic C-C bond cleavage of 3''-oxo-puerarin. Sci Rep 2023; 13:16282. [PMID: 37770535 PMCID: PMC10539306 DOI: 10.1038/s41598-023-43379-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/22/2023] [Indexed: 09/30/2023] Open
Abstract
Puerarin, daidzein C-glucoside, was known to be biotransformed to daidzein by human intestinal bacteria, which is eventually converted to (S)-equol. The metabolic pathway of puerarin to daidzein by DgpABC of Dorea sp. PUE strain was reported as puerarin (1) → 3''-oxo-puerarin (2) → daidzein (3) + hexose enediolone (C). The second reaction is the cleavage of the glycosidic C-C bond, supposedly through the quinoid intermediate (4). In this work, the glycosidic C-C bond cleavage reaction of 3''-oxo-puerarin (2) was theoretically studied by means of DFT calculation to elucidate chemical reaction mechanism, along with biochemical energetics of puerarin metabolism. It was found that bioenergetics of puerarin metabolism is slightly endergonic by 4.99 kcal/mol, mainly due to the reaction step of hexose enediolone (C) to 3''-oxo-glucose (A). The result implied that there could be additional biochemical reactions for the metabolism of hexose enediolone (C) to overcome the thermodynamic energy barrier of 4.59 kcal/mol. The computational study focused on the C-C bond cleavage of 3''-oxo-puerarin (2) found that formation of the quinoid intermediate (4) was not accessible thermodynamically, rather the reaction was initiated by the deprotonation of 2''C-H proton of 3''-oxo-puerarin (2). The 2''C-dehydro-3''-oxo-puerarin (2a2C) anionic species produced hexose enediolone (C) and 8-dehydro-daidzein anion (3a8), and the latter quickly converted to daidzein through the daidzein anion (3a7). Our study also explains why the reverse reaction of C-glycoside formation from daidzein (3) and hexose enediolone (C) is not feasible.
Collapse
Affiliation(s)
- Jongkeun Choi
- Department of Chemical Engineering, Chungwoon University, 113, Sukgol-ro, Michuhol-gu, Incheon, 22100, Republic of Korea
| | - Yongho Kim
- Department of Applied Chemistry, Institute of Applied Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Bekir Engin Eser
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000, Aarhus, Denmark
| | - Jaehong Han
- Metalloenzyme Research Group, Department of Plant Science and Technology, Chung-Ang University, 4726 Seodong-daero, Anseong, 17546, Republic of Korea.
| |
Collapse
|
28
|
Hou J, Wang Y, Zhu P, Yang N, Liang L, Yu T, Niu M, Konhauser K, Woodcroft BJ, Wang F. Taxonomic and carbon metabolic diversification of Bathyarchaeia during its coevolution history with early Earth surface environment. SCIENCE ADVANCES 2023; 9:eadf5069. [PMID: 37406125 PMCID: PMC10321748 DOI: 10.1126/sciadv.adf5069] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 06/01/2023] [Indexed: 07/07/2023]
Abstract
Bathyarchaeia, as one of the most abundant microorganisms on Earth, play vital roles in the global carbon cycle. However, our understanding of their origin, evolution, and ecological functions remains poorly constrained. Here, we present the largest dataset of Bathyarchaeia metagenome assembled genome to date and reclassify Bathyarchaeia into eight order-level units corresponding to the former subgroup system. Highly diversified and versatile carbon metabolisms were found among different orders, particularly atypical C1 metabolic pathways, indicating that Bathyarchaeia represent overlooked important methylotrophs. Molecular dating results indicate that Bathyarchaeia diverged at ~3.3 billion years, followed by three major diversifications at ~3.0, ~2.5, and ~1.8 to 1.7 billion years, likely driven by continental emergence, growth, and intensive submarine volcanism, respectively. The lignin-degrading Bathyarchaeia clade emerged at ~300 million years perhaps contributed to the sharply decreased carbon sequestration rate during the Late Carboniferous period. The evolutionary history of Bathyarchaeia potentially has been shaped by geological forces, which, in turn, affected Earth's surface environment.
Collapse
Affiliation(s)
- Jialin Hou
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yinzhao Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Pengfei Zhu
- School of Oceanography, Shanghai Jiao Tong University, Shanghai, China
| | - Na Yang
- School of Oceanography, Shanghai Jiao Tong University, Shanghai, China
| | - Lewen Liang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Tiantian Yu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Mingyang Niu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Kurt Konhauser
- Department of Earth and Atmospheric Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ben J. Woodcroft
- Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba, Australia
| | - Fengping Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- School of Oceanography, Shanghai Jiao Tong University, Shanghai, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| |
Collapse
|
29
|
Dukes HE, Tinker KA, Ottesen EA. Disentangling hindgut metabolism in the American cockroach through single-cell genomics and metatranscriptomics. Front Microbiol 2023; 14:1156809. [PMID: 37323917 PMCID: PMC10266427 DOI: 10.3389/fmicb.2023.1156809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/08/2023] [Indexed: 06/17/2023] Open
Abstract
Omnivorous cockroaches host a complex hindgut microbiota comprised of insect-specific lineages related to those found in mammalian omnivores. Many of these organisms have few cultured representatives, thereby limiting our ability to infer the functional capabilities of these microbes. Here we present a unique reference set of 96 high-quality single cell-amplified genomes (SAGs) from bacterial and archaeal cockroach gut symbionts. We additionally generated cockroach hindgut metagenomic and metatranscriptomic sequence libraries and mapped them to our SAGs. By combining these datasets, we are able to perform an in-depth phylogenetic and functional analysis to evaluate the abundance and activities of the taxa in vivo. Recovered lineages include key genera within Bacteroidota, including polysaccharide-degrading taxa from the genera Bacteroides, Dysgonomonas, and Parabacteroides, as well as a group of unclassified insect-associated Bacteroidales. We also recovered a phylogenetically diverse set of Firmicutes exhibiting a wide range of metabolic capabilities, including-but not limited to-polysaccharide and polypeptide degradation. Other functional groups exhibiting high relative activity in the metatranscriptomic dataset include multiple putative sulfate reducers belonging to families in the Desulfobacterota phylum and two groups of methanogenic archaea. Together, this work provides a valuable reference set with new insights into the functional specializations of insect gut symbionts and frames future studies of cockroach hindgut metabolism.
Collapse
Affiliation(s)
- Helen E. Dukes
- Department of Microbiology, University of Georgia, Athens, GA, United States
| | - Kara A. Tinker
- National Energy Technology Laboratory (NETL), Pittsburgh, PA, United States
| | | |
Collapse
|
30
|
Kaur A, Scott NE, Herisse M, Goddard-Borger ED, Pidot S, Williams SJ. Identification of levoglucosan degradation pathways in bacteria and sequence similarity network analysis. Arch Microbiol 2023; 205:155. [PMID: 37000297 PMCID: PMC10066097 DOI: 10.1007/s00203-023-03506-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/01/2023]
Abstract
Levoglucosan is produced in the pyrolysis of cellulose and starch, including from bushfires or the burning of biofuels, and is deposited from the atmosphere across the surface of the earth. We describe two levoglucosan degrading Paenarthrobacter spp. (Paenarthrobacter nitrojuajacolis LG01 and Paenarthrobacter histidinolovorans LG02) that were isolated from soil by metabolic enrichment using levoglucosan as the sole carbon source. Genome sequencing and proteomics analysis revealed the expression of a series of genes encoding known levoglucosan degrading enzymes, levoglucosan dehydrogenase (LGDH, LgdA), 3-keto-levoglucosan β -eliminase (LgdB1) and glucose 3-dehydrogenase (LgdC), along with an ABC transporter cassette and an associated solute binding protein. However, no homologues of 3-ketoglucose dehydratase (LgdB2) were evident, while the expressed genes contained a range of putative sugar phosphate isomerases/xylose isomerases with weak similarity to LgdB2. Sequence similarity network analysis of genome neighbours of LgdA revealed that homologues of LgdB1 and LgdC are generally conserved in a range of bacteria in the phyla Firmicutes, Actinobacteria and Proteobacteria. One group of sugar phosphate isomerase/xylose isomerase homologues (named LgdB3) was identified with limited distribution that is mutually exclusive with LgdB2, and we propose that they may fulfil a similar function. LgdB1, LgdB2 and LgdB3 adopt similar predicted 3D folds, suggesting overlapping function in processing intermediates in LG metabolism. Our findings highlight diversity within the LGDH pathway, through which bacteria utilize levoglucosan as a nutrient source.
Collapse
Affiliation(s)
- Arashdeep Kaur
- School of Chemistry, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, 3000, Australia
| | - Marion Herisse
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, 3000, Australia
| | - Ethan D Goddard-Borger
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3010, Australia
| | - Sacha Pidot
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria, 3000, Australia
| | - Spencer J Williams
- School of Chemistry, University of Melbourne, Parkville, VIC, 3010, Australia.
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
31
|
Anso I, Naegeli A, Cifuente JO, Orrantia A, Andersson E, Zenarruzabeitia O, Moraleda-Montoya A, García-Alija M, Corzana F, Del Orbe RA, Borrego F, Trastoy B, Sjögren J, Guerin ME. Turning universal O into rare Bombay type blood. Nat Commun 2023; 14:1765. [PMID: 36997505 PMCID: PMC10063614 DOI: 10.1038/s41467-023-37324-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/09/2023] [Indexed: 04/01/2023] Open
Abstract
AbstractRed blood cell antigens play critical roles in blood transfusion since donor incompatibilities can be lethal. Recipients with the rare total deficiency in H antigen, the Oh Bombay phenotype, can only be transfused with group Oh blood to avoid serious transfusion reactions. We discover FucOB from the mucin-degrading bacteria Akkermansia muciniphila as an α-1,2-fucosidase able to hydrolyze Type I, Type II, Type III and Type V H antigens to obtain the afucosylated Bombay phenotype in vitro. X-ray crystal structures of FucOB show a three-domain architecture, including a GH95 glycoside hydrolase. The structural data together with site-directed mutagenesis, enzymatic activity and computational methods provide molecular insights into substrate specificity and catalysis. Furthermore, using agglutination tests and flow cytometry-based techniques, we demonstrate the ability of FucOB to convert universal O type into rare Bombay type blood, providing exciting possibilities to facilitate transfusion in recipients/patients with Bombay phenotype.
Collapse
|
32
|
Morais MAB, Nin-Hill A, Rovira C. Glycosidase mechanisms: Sugar conformations and reactivity in endo- and exo-acting enzymes. Curr Opin Chem Biol 2023; 74:102282. [PMID: 36931022 DOI: 10.1016/j.cbpa.2023.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/19/2023] [Accepted: 02/09/2023] [Indexed: 03/17/2023]
Abstract
The enzymatic breakdown of carbohydrates plays a critical role in several biological events and enables the development of sustainable processes to obtain bioproducts and biofuels. In this scenario, the design of efficient inhibitors for glycosidases that can act as drug targets and the engineering of carbohydrate-active enzymes with tailored catalytic properties is of remarkable importance. To guide rational approaches, it is necessary to elucidate enzyme molecular mechanisms, in particular understanding how the microenvironment modulates the conformational space explored by the substrate. Computer simulations, especially those based on ab initio methods, have provided a suitable atomic description of carbohydrate conformations and catalytic reactions in several glycosidase families. In this review, we will focus on how the active-site topology (pocket or cleft) and mode of cleavage (endo or exo) can affect the catalytic mechanisms adopted by glycosidases, in particular the substrate conformations along the reaction coordinate.
Collapse
Affiliation(s)
- Mariana Abrahão Bueno Morais
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil
| | - Alba Nin-Hill
- Departament de Química Inorgànica i Orgànica & Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08028, Spain
| | - Carme Rovira
- Departament de Química Inorgànica i Orgànica & Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08028, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain.
| |
Collapse
|
33
|
Anisha GS. Biopharmaceutical applications of α-galactosidases. Biotechnol Appl Biochem 2023; 70:257-267. [PMID: 35436353 DOI: 10.1002/bab.2349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/04/2022] [Indexed: 11/06/2022]
Abstract
α-Galactosidases are exoglycosidases that are active on galactose-containing side chains in oligosaccharides, polysaccharides, glycolipids, and glycoproteins. α-Galactosidases are gaining increased interest in human medicine, especially in the enzyme replacement therapy for Fabry's disease. α-Galactosidases with regioselectivity toward α-1,3-linked galactose find application in xenotransplantation and blood group transformation. The use of α-galactosidases as a therapeutic agent in alleviating the postprandial symptoms of irritable bowel syndrome is much acclaimed. The excellent therapeutic applications of α-galactosidases have led to an upwelling of worldwide research interventions to identify novel α-galactosidases with improved catalytic efficiency. In addition to these therapeutic applications, α-galactosidases also have interesting applications in the industrial sectors like food, feed, probiotics, sugar, and paper pulp. The current review focuses on the diverse therapeutic applications of α-galactosidases and their prospects.
Collapse
Affiliation(s)
- Grace Sathyanesan Anisha
- Post-Graduate and Research Department of Zoology, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
34
|
Jajosky RP, Wu SC, Zheng L, Jajosky AN, Jajosky PG, Josephson CD, Hollenhorst MA, Sackstein R, Cummings RD, Arthur CM, Stowell SR. ABO blood group antigens and differential glycan expression: Perspective on the evolution of common human enzyme deficiencies. iScience 2023; 26:105798. [PMID: 36691627 PMCID: PMC9860303 DOI: 10.1016/j.isci.2022.105798] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Enzymes catalyze biochemical reactions and play critical roles in human health and disease. Enzyme variants and deficiencies can lead to variable expression of glycans, which can affect physiology, influence predilection for disease, and/or directly contribute to disease pathogenesis. Although certain well-characterized enzyme deficiencies result in overt disease, some of the most common enzyme deficiencies in humans form the basis of blood groups. These carbohydrate blood groups impact fundamental areas of clinical medicine, including the risk of infection and severity of infectious disease, bleeding risk, transfusion medicine, and tissue/organ transplantation. In this review, we examine the enzymes responsible for carbohydrate-based blood group antigen biosynthesis and their expression within the human population. We also consider the evolutionary selective pressures, e.g. malaria, that may account for the variation in carbohydrate structures and the implications of this biology for human disease.
Collapse
Affiliation(s)
- Ryan Philip Jajosky
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Biconcavity Inc, Lilburn, GA, USA
| | - Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Leon Zheng
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Audrey N. Jajosky
- University of Rochester Medical Center, Department of Pathology and Laboratory Medicine, West Henrietta, NY, USA
| | | | - Cassandra D. Josephson
- Cancer and Blood Disorders Institute and Blood Bank/Transfusion Medicine Division, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
- Departments of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marie A. Hollenhorst
- Department of Pathology and Department of Medicine, Stanford University, Stanford, CA, USA
| | - Robert Sackstein
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Connie M. Arthur
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Sean R. Stowell
- Joint Program in Transfusion Medicine, Brigham and Women’s Hospital, Harvard Medical School, 630E New Research Building, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
35
|
Ren Y, Eronen V, Blomster Andberg M, Koivula A, Hakulinen N. Structure and function of aldopentose catabolism enzymes involved in oxidative non-phosphorylative pathways. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:147. [PMID: 36578086 PMCID: PMC9795676 DOI: 10.1186/s13068-022-02252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Platform chemicals and polymer precursors can be produced via enzymatic pathways starting from lignocellulosic waste materials. The hemicellulose fraction of lignocellulose contains aldopentose sugars, such as D-xylose and L-arabinose, which can be enzymatically converted into various biobased products by microbial non-phosphorylated oxidative pathways. The Weimberg and Dahms pathways convert pentose sugars into α-ketoglutarate, or pyruvate and glycolaldehyde, respectively, which then serve as precursors for further conversion into a wide range of industrial products. In this review, we summarize the known three-dimensional structures of the enzymes involved in oxidative non-phosphorylative pathways of pentose catabolism. Key structural features and reaction mechanisms of a diverse set of enzymes responsible for the catalytic steps in the reactions are analysed and discussed.
Collapse
Affiliation(s)
- Yaxin Ren
- grid.9668.10000 0001 0726 2490Department of Chemistry, University of Eastern Finland, 111, 80101 Joensuu, Finland
| | - Veikko Eronen
- grid.9668.10000 0001 0726 2490Department of Chemistry, University of Eastern Finland, 111, 80101 Joensuu, Finland
| | | | - Anu Koivula
- grid.6324.30000 0004 0400 1852VTT Technical Research Centre of Finland Ltd, Espoo, Finland
| | - Nina Hakulinen
- grid.9668.10000 0001 0726 2490Department of Chemistry, University of Eastern Finland, 111, 80101 Joensuu, Finland
| |
Collapse
|
36
|
Rahman MA, Heme UH, Parvez MAK. In silico functional annotation of hypothetical proteins from the Bacillus paralicheniformis strain Bac84 reveals proteins with biotechnological potentials and adaptational functions to extreme environments. PLoS One 2022; 17:e0276085. [PMID: 36228026 PMCID: PMC9560612 DOI: 10.1371/journal.pone.0276085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/28/2022] [Indexed: 11/26/2022] Open
Abstract
Members of the Bacillus genus are industrial cell factories due to their capacity to secrete significant quantities of biomolecules with industrial applications. The Bacillus paralicheniformis strain Bac84 was isolated from the Red Sea and it shares a close evolutionary relationship with Bacillus licheniformis. However, a significant number of proteins in its genome are annotated as functionally uncharacterized hypothetical proteins. Investigating these proteins' functions may help us better understand how bacteria survive extreme environmental conditions and to find novel targets for biotechnological applications. Therefore, the purpose of our research was to functionally annotate the hypothetical proteins from the genome of B. paralicheniformis strain Bac84. We employed a structured in-silico approach incorporating numerous bioinformatics tools and databases for functional annotation, physicochemical characterization, subcellular localization, protein-protein interactions, and three-dimensional structure determination. Sequences of 414 hypothetical proteins were evaluated and we were able to successfully attribute a function to 37 hypothetical proteins. Moreover, we performed receiver operating characteristic analysis to assess the performance of various tools used in this present study. We identified 12 proteins having significant adaptational roles to unfavorable environments such as sporulation, formation of biofilm, motility, regulation of transcription, etc. Additionally, 8 proteins were predicted with biotechnological potentials such as coenzyme A biosynthesis, phenylalanine biosynthesis, rare-sugars biosynthesis, antibiotic biosynthesis, bioremediation, and others. Evaluation of the performance of the tools showed an accuracy of 98% which represented the rationality of the tools used. This work shows that this annotation strategy will make the functional characterization of unknown proteins easier and can find the target for further investigation. The knowledge of these hypothetical proteins' potential functions aids B. paralicheniformis strain Bac84 in effectively creating a new biotechnological target. In addition, the results may also facilitate a better understanding of the survival mechanisms in harsh environmental conditions.
Collapse
Affiliation(s)
- Md. Atikur Rahman
- Institute of Microbiology, Friedrich Schiller University Jena, Thuringia, Germany
| | - Uzma Habiba Heme
- Faculty of Biological Sciences, Friedrich Schiller University Jena, Thuringia, Germany
| | | |
Collapse
|
37
|
Petazzi P, Miquel‐Serra L, Huertas S, González C, Boto N, Muñiz‐Diaz E, Menéndez P, Sevilla A, Nogués N. ABO gene editing for the conversion of blood type A to universal type O in Rh null donor-derived human-induced pluripotent stem cells. Clin Transl Med 2022; 12:e1063. [PMID: 36281739 PMCID: PMC9593258 DOI: 10.1002/ctm2.1063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/13/2022] [Accepted: 09/06/2022] [Indexed: 01/28/2023] Open
Abstract
The limited availability of red cells with extremely rare blood group phenotypes is one of the global challenges in transfusion medicine that has prompted the search for alternative self-renewable pluripotent cell sources for the in vitro generation of red cells with rare blood group types. One such phenotype is the Rhnull , which lacks all the Rh antigens on the red cell membrane and represents one of the rarest blood types in the world with only a few active blood donors available worldwide. Rhnull red cells are critical for the transfusion of immunized patients carrying the same phenotype, besides its utility in the diagnosis of Rh alloimmunization when a high-prevalence Rh specificity is suspected in a patient or a pregnant woman. In both scenarios, the potential use of human-induced pluripotent stem cell (hiPSC)-derived Rhnull red cells is also dependent on ABO compatibility. Here, we present a CRISPR/Cas9-mediated ABO gene edition strategy for the conversion of blood type A to universal type O, which we have applied to an Rhnull donor-derived hiPSC line, originally carrying blood group A. This work provides a paradigmatic example of an approach potentially applicable to other hiPSC lines derived from rare blood donors not carrying blood type O.
Collapse
Affiliation(s)
- Paolo Petazzi
- Josep Carreras Leukemia Research InstituteBarcelonaSpain
| | - Laia Miquel‐Serra
- Immunohematology LaboratoryBarcelonaSpain
- Transfusional medicine. Vall d'Hebron Research Institute (VHIR)BarcelonaSpain
| | - Sergio Huertas
- Immunohematology LaboratoryBarcelonaSpain
- Transfusional medicine. Vall d'Hebron Research Institute (VHIR)BarcelonaSpain
| | - Cecilia González
- Immunohematology LaboratoryBarcelonaSpain
- Transfusional medicine. Vall d'Hebron Research Institute (VHIR)BarcelonaSpain
| | - Neus Boto
- Immunohematology LaboratoryBarcelonaSpain
| | - Eduardo Muñiz‐Diaz
- Immunohematology LaboratoryBarcelonaSpain
- Transfusional medicine. Vall d'Hebron Research Institute (VHIR)BarcelonaSpain
- Department of MedicineUniversitat Autònoma de Barcelona (UAB)BarcelonaSpain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research InstituteBarcelonaSpain
- Department of Biomedicine, School of MedicineUniversity of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Cáncer‐CIBER‐ONCInstituto de Salud Carlos IIIBarcelonaSpain
- Red Española de Terapias Avanzadas (TERAV)Instituto de Salud Carlos III (RICORS, RD21/0017/0029)
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Ana Sevilla
- Department of Cell BiologyPhysiology and Immunology, Faculty of Biology, University of BarcelonaBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
| | - Núria Nogués
- Immunohematology LaboratoryBarcelonaSpain
- Transfusional medicine. Vall d'Hebron Research Institute (VHIR)BarcelonaSpain
- Department of MedicineUniversitat Autònoma de Barcelona (UAB)BarcelonaSpain
| |
Collapse
|
38
|
Ristovska E, Bojadjieva ТM, Velkova Е, Dimceva АH, Todorovski B, Tashkovska M, Rastvorceva RG, Bosevski M. Rare Blood Groups in ABO, Rh, Kell Systems - Biological and Clinical Significance. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 2022; 43:77-87. [PMID: 35843921 DOI: 10.2478/prilozi-2022-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Background: The frequency of ABO, Rh and Kell blood group antigens differs among populations of different ethnic ancestry. There are low-frequency antigens (<1%) and high-frequency antigens (>90%). A rare blood group is defined as the absence of a high-frequency antigen in the general population, as well as absence of multiple frequent antigens within a single or multiple blood group systems. Aim: To perform red blood cell typing and to calculate the antigen and phenotype frequencies, in order to identify rare blood group donors within the clinically most important АВО, Rh and Kell systems. Material and Methods: АВО, Rh (D, C, E, c, e) and Kell (K) antigen typing was performed using specific monoclonal sera and microplate technique, while Cellano (k) typing was performed with a monoclonal anti-k, antihuman globulin and column agglutination technique. Weak ABO subgroups were determined using the absorption elution method or molecular genotyping (PCR-SSP). Results: ABO antigen frequency is: A (40.89%), O (34.22%), B (16.97%), AB (7.92%) and weak ABO subgroups (0, 009 %). The established genotypes were AxO1 (0, 0026%) and AxB (0, 001%). Rh antigen frequency is: D (85.79%), C (71.7%), c (76.0%), E (26.0%) and е (97.95%). The most common Rh pheno-type is the DCcee (32.7%) while the rarest phenotype is the DCCEE phenotype (0. 003%). The prevalence of K and k antigen is 7.5% and 99.94%, respectively. The frequency of the rare phenotype K+k- is 0.06%. Conclusion: Large scale phenotyping of blood group antigens enables the identification of blood donors with rare blood groups for patients with rare phenotypes or with antibodies to high-frequency antigens and to frequent antigens within one or more blood group systems.
Collapse
Affiliation(s)
- Elena Ristovska
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | | | - Еmilija Velkova
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | - Аnita Hristova Dimceva
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | - Bojan Todorovski
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | - Marija Tashkovska
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | - Rada Grubovic Rastvorceva
- Institute for transfusion medicine, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| | - Marijan Bosevski
- Clinic of cardiology, St Cyril and Methodius University, Medical Faculty, Skopje, RN Macedonia
| |
Collapse
|
39
|
Ameri R, García JL, Derenfed AB, Pradel N, Neifar S, Mhiri S, Mezghanni M, Jaouadi NZ, Barriuso J, Bejar S. Genome sequence and Carbohydrate Active Enzymes (CAZymes) repertoire of the thermophilic Caldicoprobacter algeriensis TH7C1 T. Microb Cell Fact 2022; 21:91. [PMID: 35598016 PMCID: PMC9124407 DOI: 10.1186/s12934-022-01818-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/05/2022] [Indexed: 12/31/2022] Open
Abstract
Background Omics approaches are widely applied in the field of biology for the discovery of potential CAZymes including whole genome sequencing. The aim of this study was to identify protein encoding genes including CAZymes in order to understand glycans-degrading machinery in the thermophilic Caldicoprobacter algeriensis TH7C1T strain. Results Caldicoprobacter algeriensis TH7C1T is a thermophilic anaerobic bacterium belonging to the Firmicutes phylum, which grows between the temperatures of 55 °C and 75 °C. Next generation sequencing using Illumina technology was performed on the C. algeriensis strain resulting in 45 contigs with an average GC content of 44.9% and a total length of 2,535,023 bp. Genome annotation reveals 2425 protein-coding genes with 97 ORFs coding CAZymes. Many glycoside hydrolases, carbohydrate esterases and glycosyltransferases genes were found linked to genes encoding oligosaccharide transporters and transcriptional regulators; suggesting that CAZyme encoding genes are organized in clusters involved in polysaccharides degradation and transport. In depth analysis of CAZomes content in C. algeriensis genome unveiled 33 CAZyme gene clusters uncovering new enzyme combinations targeting specific substrates. Conclusions This study is the first targeting CAZymes repertoire of C. algeriensis, it provides insight to the high potential of identified enzymes for plant biomass degradation and their biotechnological applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01818-0.
Collapse
Affiliation(s)
- Rihab Ameri
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia
| | - José Luis García
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), C/ Ramiro de Maeztu 9, 28040, Madrid, Spain.,IBISBA_ES_CSIC_Cell Factory_MM, Madrid, Spain
| | - Amel Bouanane Derenfed
- Laboratoire de Biologie Cellulaire et Moléculaire (Équipe de Microbiologie), Université des Sciences et de la Technologie Houari Boumédiènne, Bab Ezzouar, Algiers, Algeria
| | - Nathalie Pradel
- Université de Toulon, CNRS, IRD, MIO, Aix Marseille Univ, Marseille, France
| | - Sawssan Neifar
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia
| | - Sonia Mhiri
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia
| | - Monia Mezghanni
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia
| | - Nadia Zaraî Jaouadi
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia
| | - Jorge Barriuso
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), C/ Ramiro de Maeztu 9, 28040, Madrid, Spain.,IBISBA_ES_CSIC_Cell Factory_MM, Madrid, Spain
| | - Samir Bejar
- Laboratory of Microbial Biotechnology, Enzymatic and Biomolecules, Centre of Biotechnology of Sfax (CBS), University of Sfax, Sidi Mansour Road Km 6, P.O. Box 1177, 3018, Sfax, Tunisia.
| |
Collapse
|
40
|
Wardman JF, Bains RK, Rahfeld P, Withers SG. Carbohydrate-active enzymes (CAZymes) in the gut microbiome. Nat Rev Microbiol 2022; 20:542-556. [PMID: 35347288 DOI: 10.1038/s41579-022-00712-1] [Citation(s) in RCA: 253] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Abstract
The 1013-1014 microorganisms present in the human gut (collectively known as the human gut microbiota) dedicate substantial percentages of their genomes to the degradation and uptake of carbohydrates, indicating the importance of this class of molecules. Carbohydrates function not only as a carbon source for these bacteria but also as a means of attachment to the host, and a barrier to infection of the host. In this Review, we focus on the diversity of carbohydrate-active enzymes (CAZymes), how gut microorganisms use them for carbohydrate degradation, the different chemical mechanisms of these CAZymes and the roles that these microorganisms and their CAZymes have in human health and disease. We also highlight examples of how enzymes from this treasure trove have been used in manipulation of the microbiota for improved health and treatment of disease, in remodelling the glycans on biopharmaceuticals and in the potential production of universal O-type donor blood.
Collapse
Affiliation(s)
- Jacob F Wardman
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rajneesh K Bains
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter Rahfeld
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen G Withers
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada. .,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
41
|
Zhang M, Guo H, Xia D, Dong Z, Liu X, Zhao W, Jia J, Yin X. Metagenomic insight of corn straw conditioning on substrates metabolism during coal anaerobic fermentation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 808:152220. [PMID: 34890652 DOI: 10.1016/j.scitotenv.2021.152220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/14/2021] [Accepted: 12/02/2021] [Indexed: 06/13/2023]
Abstract
Increasing methane production from anaerobic digestion of coal is challenging. This study shows that the combined fermentation of coal and corn straw greatly enriched the substrates available to microorganisms. This was mainly manifested in the increased types and abundance of organic matter in the fermentation liquid, which enhanced methane production by 61%. Metagenomic analysis showed that the addition of corn straw enriched the abundance of Methanosarcina in the combined fermentation system and promoted the complementary advantages of the microorganisms. At the same time, the abundance of genes that convert glucose into acetic acid (K00927, K01689, K01905, etc.) in the combined fermentation system increased, which is conducive to acidification process and biomethane production. In addition, there were the two key methanogenic pathways, namely aceticlastic (57.1%-63.5%) and hydrogenotrophic (23.4%-25.1%) methanogenesis, identified in the single coal fermentation system and the combined coal and corn straw fermentation system. Combined fermentation enhanced the hydrogenotrophic and methylotrophic methanogenic pathways by increasing the gene abundance of K00200 (methane production from CO2 and oxidation of coenzyme M to CO2), K00440 (participates in the binding to other known physiological receptors with hydrogen as a donor), and K00577 (methyltransferase).
Collapse
Affiliation(s)
- Minglu Zhang
- School of Energy Science and Engineering, Henan Polytechnic University, Jiaozuo 454000, China
| | - Hongyu Guo
- School of Energy Science and Engineering, Henan Polytechnic University, Jiaozuo 454000, China.
| | - Daping Xia
- School of Energy Science and Engineering, Henan Polytechnic University, Jiaozuo 454000, China.
| | - Zhiwei Dong
- School of Energy Science and Engineering, Henan Polytechnic University, Jiaozuo 454000, China
| | - Xile Liu
- School of Geoscience and Surveying Engineering, China University of Mining &Technology, Beijing 100083, China
| | - Weizhong Zhao
- Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo 454000, China
| | - Jianbo Jia
- College of Chemistry and Chemical Engineering, Henan Polytechnic University, Jiaozuo 454000, China
| | - Xiangju Yin
- School of Emergency Management, Henan Polytechnic University, Jiaozuo 454000, China.
| |
Collapse
|
42
|
Wang A, Ribeiro RVP, Ali A, Brambate E, Abdelnour-Berchtold E, Michaelsen V, Zhang Y, Rahfeld P, Moon H, Gokhale H, Gazzalle A, Pal P, Liu M, Waddell TK, Cserti-Gazdewich C, Tinckam K, Kizhakkedathu JN, West L, Keshavjee S, Withers SG, Cypel M. Ex vivo enzymatic treatment converts blood type A donor lungs into universal blood type lungs. Sci Transl Med 2022; 14:eabm7190. [PMID: 35171649 DOI: 10.1126/scitranslmed.abm7190] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Donor organ allocation is dependent on ABO matching, restricting the opportunity for some patients to receive a life-saving transplant. The enzymes FpGalNAc deacetylase and FpGalactosaminidase, used in combination, have been described to effectively convert group A (ABO-A) red blood cells (RBCs) to group O (ABO-O). Here, we study the safety and preclinical efficacy of using these enzymes to remove A antigen (A-Ag) from human donor lungs using ex vivo lung perfusion (EVLP). First, the ability of these enzymes to remove A-Ag in organ perfusate solutions was examined on five human ABO-A1 RBC samples and three human aortae after static incubation. The enzymes removed greater than 99 and 90% A-Ag from RBCs and aortae, respectively, at concentrations as low as 1 μg/ml. Eight ABO-A1 human lungs were then treated by EVLP. Baseline analyses of A-Ag in lungs revealed expression predominantly in the endothelial and epithelial cells. EVLP of lungs with enzyme-containing perfusate removed over 97% of endothelial A-Ag within 4 hours. No treatment-related acute lung toxicity was observed. An ABO-incompatible transplant was then simulated with an ex vivo model of antibody-mediated rejection using ABO-O plasma as the surrogate for the recipient circulation using three donor lungs. The treatment of donor lungs minimized antibody binding, complement deposition, and antibody-mediated injury as compared with control lungs. These results show that depletion of donor lung A-Ag can be achieved with EVLP treatment. This strategy has the potential to expand ABO-incompatible lung transplantation and lead to improvements in fairness of organ allocation.
Collapse
Affiliation(s)
- Aizhou Wang
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Rafaela V P Ribeiro
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Aadil Ali
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Edson Brambate
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Etienne Abdelnour-Berchtold
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Vinicius Michaelsen
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Yu Zhang
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Peter Rahfeld
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Haisle Moon
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hemant Gokhale
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Anajara Gazzalle
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada
| | - Prodipto Pal
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada.,Departments of Surgery, Medicine and Physiology and Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, ON M5T 1P5, Canada
| | - Thomas K Waddell
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | | | - Kathryn Tinckam
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON M5S 1A8, Canada.,Department of Medicine, University Health Network and University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, Life Science Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lori West
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada.,Canadian Donation and Transplantation Research Program, Edmonton AB T6G 1C9, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, ON M5G 1L7, Canada.,Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
43
|
Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, Sanders JG, Valsta L, Brożyńska M, Zhu Q, Tripathi A, Vázquez-Baeza Y, Loomba R, Cheng S, Jain M, Niiranen T, Lahti L, Knight R, Salomaa V, Inouye M, Méric G. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet 2022; 54:134-142. [PMID: 35115689 PMCID: PMC9883041 DOI: 10.1038/s41588-021-00991-z] [Citation(s) in RCA: 248] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/19/2021] [Indexed: 01/31/2023]
Abstract
Human genetic variation affects the gut microbiota through a complex combination of environmental and host factors. Here we characterize genetic variations associated with microbial abundances in a single large-scale population-based cohort of 5,959 genotyped individuals with matched gut microbial metagenomes, and dietary and health records (prevalent and follow-up). We identified 567 independent SNP-taxon associations. Variants at the LCT locus associated with Bifidobacterium and other taxa, but they differed according to dairy intake. Furthermore, levels of Faecalicatena lactaris associated with ABO, and suggested preferential utilization of secreted blood antigens as energy source in the gut. Enterococcus faecalis levels associated with variants in the MED13L locus, which has been linked to colorectal cancer. Mendelian randomization analysis indicated a potential causal effect of Morganella on major depressive disorder, consistent with observational incident disease analysis. Overall, we identify and characterize the intricate nature of host-microbiota interactions and their association with disease.
Collapse
Affiliation(s)
- Youwen Qin
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Yang Liu
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Alex Tokolyi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Jon G Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
- Cornell Institute for Host-Microbe Interaction and Disease, Cornell University, Ithaca, NY, USA
| | - Liisa Valsta
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Marta Brożyńska
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Qiyun Zhu
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anupriya Tripathi
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science & Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohit Jain
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science & Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia.
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus & University of Cambridge, Cambridge, UK.
- The Alan Turing Institute, London, UK.
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
44
|
Wu H, Crost EH, Owen CD, van Bakel W, Martínez Gascueña A, Latousakis D, Hicks T, Walpole S, Urbanowicz PA, Ndeh D, Monaco S, Sánchez Salom L, Griffiths R, Reynolds RS, Colvile A, Spencer DIR, Walsh M, Angulo J, Juge N. The human gut symbiont Ruminococcus gnavus shows specificity to blood group A antigen during mucin glycan foraging: Implication for niche colonisation in the gastrointestinal tract. PLoS Biol 2021; 19:e3001498. [PMID: 34936658 PMCID: PMC8730463 DOI: 10.1371/journal.pbio.3001498] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/05/2022] [Accepted: 11/26/2021] [Indexed: 12/24/2022] Open
Abstract
The human gut symbiont Ruminococcus gnavus displays strain-specific repertoires of glycoside hydrolases (GHs) contributing to its spatial location in the gut. Sequence similarity network analysis identified strain-specific differences in blood-group endo-β-1,4-galactosidase belonging to the GH98 family. We determined the substrate and linkage specificities of GH98 from R. gnavus ATCC 29149, RgGH98, against a range of defined oligosaccharides and glycoconjugates including mucin. We showed by HPAEC-PAD and LC-FD-MS/MS that RgGH98 is specific for blood group A tetrasaccharide type II (BgA II). Isothermal titration calorimetry (ITC) and saturation transfer difference (STD) NMR confirmed RgGH98 affinity for blood group A over blood group B and H antigens. The molecular basis of RgGH98 strict specificity was further investigated using a combination of glycan microarrays, site-directed mutagenesis, and X-ray crystallography. The crystal structures of RgGH98 in complex with BgA trisaccharide (BgAtri) and of RgGH98 E411A with BgA II revealed a dedicated hydrogen network of residues, which were shown by site-directed mutagenesis to be critical to the recognition of the BgA epitope. We demonstrated experimentally that RgGH98 is part of an operon of 10 genes that is overexpresssed in vitro when R. gnavus ATCC 29149 is grown on mucin as sole carbon source as shown by RNAseq analysis and RT-qPCR confirmed RgGH98 expression on BgA II growth. Using MALDI-ToF MS, we showed that RgGH98 releases BgAtri from mucin and that pretreatment of mucin with RgGH98 confered R. gnavus E1 the ability to grow, by enabling the E1 strain to metabolise BgAtri and access the underlying mucin glycan chain. These data further support that the GH repertoire of R. gnavus strains enable them to colonise different nutritional niches in the human gut and has potential applications in diagnostic and therapeutics against infection.
Collapse
Affiliation(s)
- Haiyang Wu
- Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - C David Owen
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | | | | | | | - Thomas Hicks
- University of East Anglia, Norwich, United Kingdom
| | | | | | - Didier Ndeh
- Quadram Institute Bioscience, Norwich, United Kingdom
| | | | | | | | | | - Anna Colvile
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | | | - Martin Walsh
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | - Jesus Angulo
- University of East Anglia, Norwich, United Kingdom
- Universidad de Sevilla and Instituto de Investigaciones Químicas, Sevilla, Spain
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich, United Kingdom
- * E-mail:
| |
Collapse
|
45
|
Moorthy NSHN, Brás NF, Ramos MJ, Fernandes PA. Structure based virtual screening of natural product molecules as glycosidase inhibitors. In Silico Pharmacol 2021; 9:56. [PMID: 34722112 DOI: 10.1007/s40203-021-00115-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/01/2021] [Indexed: 11/30/2022] Open
Abstract
Objective of the present investigation comprised of the application of in silico methods to discover novel natural product (NP) based potential inhibitors for carbohydrate mediated diseases. Structure based drug design studies (molecular docking and structure based pharmacophore analysis) were carried out on a series of natural product compounds to identify significant bioactive molecules to inhibit α-mannosidase (I and II) and β-galactosidase enzymes. Furthermore, protein ligand interaction fingerprint analysis, molecular dynamics simulations and molecular access system (MACCS) fingerprint analysis were performed to understand the binding behaviors of the studied molecules. The results derived from these analyses showed that the identified compounds exhibit significant binding interactions with the active site residues. The compounds, NP-51, NP-81 and NP-165 have shown significant docking score against the studied enzymes (α-mannosidases-I, α-mannosidases-II and β-galactosidases). The fingerprint studies showed that the presence of rings (aromatic or aliphatic) with sulfur atoms, nitrogen atoms, methyl groups, etc. have favorable effects on the α-mannosidase II inhibitory activity. However, the presence of halogen atoms substituted in the molecules have reduced inhibitory ability against α-mannosidase II. The compound, NP-165 has significant activity against both enzymes (α-mannosidases and β-galactosidases). These studies accomplished that the compounds identified through in silico methodologies can be used to develop semisynthetic derivatives of the glycosidase inhibitors and can be screened for the treatment of different carbohydrate mediated diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-021-00115-9.
Collapse
Affiliation(s)
- N S Hari Narayana Moorthy
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, MP 484887 India.,UCIBIO, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 6874169-007 Porto, Portugal
| | - Natércia F Brás
- UCIBIO, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 6874169-007 Porto, Portugal
| | - Maria J Ramos
- UCIBIO, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 6874169-007 Porto, Portugal
| | - Pedro A Fernandes
- UCIBIO, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 6874169-007 Porto, Portugal
| |
Collapse
|
46
|
Quirke JCK, Crich D. GH47 and Other Glycoside Hydrolases Catalyze Glycosidic Bond Cleavage with the Assistance of Substrate Super-arming at the Transition State. ACS Catal 2021; 11:10308-10315. [PMID: 34777906 PMCID: PMC8579916 DOI: 10.1021/acscatal.1c02750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Super-armed glycosyl donors, whose substituents are predominantly held in pseudoaxial positions, exhibit strongly increased reactivity in glycosylation through significant stabilization of oxocarbenium-like transition states. Examination of X-ray crystal structures reveals that the GH47 family of glycoside hydrolases have evolved so as to distort their substrates away from the ground state conformation in such a manner as to present multiple C-O bonds in pseudoaxial positions and so benefit from conformational super-arming of their substrates, thereby enhancing catalysis. Through analysis of literature mutagenic studies, we show that a suitably placed aromatic residue in GHs 6 and 47 sterically enforces super-armed conformations on their substrates. GH families 45, 81, and 134 on the other hand impose conformational super-arming on their substrates, by maintaining the more active ring conformation through hydrogen bonding rather than steric interactions. The recognition of substrate super-arming by select GH families provides a further parallel with synthetic carbohydrate chemistry and nature and opens further avenues for the design of improved glycosidase inhibitors.
Collapse
Affiliation(s)
- Jonathan C K Quirke
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA 30602, USA
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, GA 30602, USA
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - David Crich
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 250 West Green Street, Athens, GA 30602, USA
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, GA 30602, USA
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
47
|
Structural basis of catalysis and substrate recognition by the NAD(H)-dependent α-d-glucuronidase from the glycoside hydrolase family 4. Biochem J 2021; 478:943-959. [PMID: 33565573 DOI: 10.1042/bcj20200824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 11/17/2022]
Abstract
Members of the glycoside hydrolase family 4 (GH4) employ an unusual glycosidic bond cleavage mechanism utilizing NAD(H) and a divalent metal ion, under reducing conditions. These enzymes act upon a diverse range of glycosides, and unlike most other GH families, homologs here are known to accommodate both α- and β-anomeric specificities within the same active site. Here, we report the catalytic properties and the crystal structures of TmAgu4B, an α-d-glucuronidase from the hyperthermophile Thermotoga maritima. The structures in three different states include the apo form, the NADH bound holo form, and the ternary complex with NADH and the reaction product d-glucuronic acid, at 2.15, 1.97 and 1.85 Å resolutions, respectively. These structures reveal the step-wise route of conformational changes required in the active site to achieve the catalytically competent state, and illustrate the direct role of residues that determine the reaction mechanism. Furthermore, a structural transition of a helical region in the active site to a turn geometry resulting in the rearrangement of a unique arginine residue governs the exclusive glucopyranosiduronic acid recognition in TmAgu4B. Mutational studies show that modifications of the glycone binding site geometry lead to catalytic failure and indicate overlapping roles of specific residues in catalysis and substrate recognition. The data highlight hitherto unreported molecular features and associated active site dynamics that determine the structure-function relationships within the unique GH4 family.
Collapse
|
48
|
Jie Z, Liang S, Ding Q, Li F, Tang S, Wang D, Lin Y, Chen P, Cai K, Qiu X, Li Q, Liao Y, Zhou D, Lian H, Zuo Y, Chen X, Rao W, Ren Y, Wang Y, Zi J, Wang R, Zhou H, Lu H, Wang X, Zhang W, Zhang T, Xiao L, Zong Y, Liu W, Yang H, Wang J, Hou Y, Liu X, Kristiansen K, Zhong H, Jia H, Xu X. A transomic cohort as a reference point for promoting a healthy human gut microbiome. MEDICINE IN MICROECOLOGY 2021. [DOI: 10.1016/j.medmic.2021.100039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
49
|
Müller M, Calvert M, Hottmann I, Kluj RM, Teufel T, Balbuchta K, Engelbrecht A, Selim KA, Xu Q, Borisova M, Titz A, Mayer C. The exo-β-N-acetylmuramidase NamZ from Bacillus subtilis is the founding member of a family of exo-lytic peptidoglycan hexosaminidases. J Biol Chem 2021; 296:100519. [PMID: 33684445 PMCID: PMC8054146 DOI: 10.1016/j.jbc.2021.100519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/27/2021] [Accepted: 03/04/2021] [Indexed: 11/11/2022] Open
Abstract
Endo-β-N-acetylmuramidases, commonly known as lysozymes, are well-characterized antimicrobial enzymes that catalyze an endo-lytic cleavage of peptidoglycan; i.e., they hydrolyze the β-1,4-glycosidic bonds connecting N-acetylmuramic acid (MurNAc) and N-acetylglucosamine (GlcNAc). In contrast, little is known about exo-β-N-acetylmuramidases, which catalyze an exo-lytic cleavage of β-1,4-MurNAc entities from the non-reducing ends of peptidoglycan chains. Such an enzyme was identified earlier in the bacterium Bacillus subtilis, but the corresponding gene has remained unknown so far. We now report that ybbC of B. subtilis, renamed namZ, encodes the reported exo-β-N-acetylmuramidase. A ΔnamZ mutant accumulated specific cell wall fragments and showed growth defects under starvation conditions, indicating a role of NamZ in cell wall turnover and recycling. Recombinant NamZ protein specifically hydrolyzed the artificial substrate para-nitrophenyl β-MurNAc and the peptidoglycan-derived disaccharide MurNAc-β-1,4-GlcNAc. Together with the exo-β-N-acetylglucosaminidase NagZ and the exo-muramoyl-l-alanine amidase AmiE, NamZ degraded intact peptidoglycan by sequential hydrolysis from the non-reducing ends. A structure model of NamZ, built on the basis of two crystal structures of putative orthologs from Bacteroides fragilis, revealed a two-domain structure including a Rossmann-fold-like domain that constitutes a unique glycosidase fold. Thus, NamZ, a member of the DUF1343 protein family of unknown function, is now classified as the founding member of a new family of glycosidases (CAZy GH171; www.cazy.org/GH171.html). NamZ-like peptidoglycan hexosaminidases are mainly present in the phylum Bacteroidetes and less frequently found in individual genomes within Firmicutes (Bacilli, Clostridia), Actinobacteria, and γ-proteobacteria.
Collapse
Affiliation(s)
- Maraike Müller
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Matthew Calvert
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, Saarland University, Saarbrücken, Germany
| | - Isabel Hottmann
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Robert Maria Kluj
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Tim Teufel
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Katja Balbuchta
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Alicia Engelbrecht
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Khaled A Selim
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany; Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Division, National Research Center, Giza, Egypt
| | - Qingping Xu
- GM/CA @ APS, Argonne National Laboratory, Lemont, Illinois, USA
| | - Marina Borisova
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany; Department of Chemistry, Saarland University, Saarbrücken, Germany
| | - Christoph Mayer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
50
|
Li B, Cui Y, Wang X, Tang R. Novel nanomaterial-organism hybrids with biomedical potential. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1706. [PMID: 33644977 DOI: 10.1002/wnan.1706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/29/2022]
Abstract
Instinctive hierarchically biomineralized structures of various organisms, such as eggs, algae, and magnetotactic bacteria, afford extra protection and distinct performance, which endow fragile organisms with a tenacious ability to adapt and survive. However, spontaneous formation of hybrid materials is difficult for most organisms in nature. Rapid development of chemistry and materials science successfully obtained the combinations of organisms with nanomaterials by biomimetic mineralization thus demonstrating the reproduction of the structures and functions and generation of novel functions that organisms do not possess. The rational design of biomaterial-organism hybridization can control biological recognition, interactions, and metabolism of the organisms. Thus, nanomaterial-organism hybrids represent a next generation of organism engineering with great potential biomedical applications. This review summarizes recent advances in material-directed organism engineering and is mainly focused on biomimetic mineralization technologies and their outstanding biomedical applications. Three representative types of biomimetic mineralization are systematically introduced, including external mineralization, internal mineralization, and genetic engineering mineralization. The methods involving hybridization of nanomaterials and organisms based on biomimetic mineralization strategies are described. These strategies resulted in applications of various nanomaterial-organism hybrids with multiplex functions in cell engineering, cancer treatment, and vaccine improvement. Unlike classical biological approaches, this material-based bioregulation is universal, effective, and inexpensive. In particular, instead of traditional medical solutions, the integration of nanomaterials and organisms may exploit novel strategies to solve current biomedical problems. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Benke Li
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yihao Cui
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China.,Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|