1
|
Marwan-Abdelbaset E, Samy-Kamal M, Tan D, Lu X. Microbial production of hyaluronic acid: The current advances, engineering strategies and trends. J Biotechnol 2025; 403:52-72. [PMID: 40154620 DOI: 10.1016/j.jbiotec.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/27/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Hyaluronic acid (HA) is a versatile biomolecule with applications in medicine, cosmetics, and pharmaceuticals. While traditionally extracted from animal tissues, HA is now predominantly produced through microbial fermentation. Microbial fermentation using strains such as Streptococcus zooepidemicus, Corynebacterium glutamicum, and Bacillus subtilis offers a more scalable and sustainable alternative to chemical and animal extraction methods. Recent studies reveal promising yields from engineered strains of Corynebacterium glutamicum and Bacillus subtilis, utilizing advanced metabolic and genetic techniques. Recent advancements in genetic and metabolic engineering, as well as synthetic biology, have addressed some challenges related to molecular weight, viscosity, and by-product formation. This review focuses on the microbial production of HA using engineered strains, encompassing producer organisms, metabolic engineering strategies, industrial-scale production, and key factors influencing molecular weight. Furthermore, it addresses the challenges and potential solutions associated with HA production. Additional research is necessary to develop more efficient and robust engineered strains that exhibit resistance to contamination and can utilize low-cost substrates, such as Pseudomonas putida and Halomonas spp. By overcoming these challenges, researchers can advance the industrial production of HA and expand its applications, thereby contributing to the growth of the HA market.
Collapse
Affiliation(s)
- Ehab Marwan-Abdelbaset
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt
| | - Mohamed Samy-Kamal
- Department of Marine Sciences and Applied Biology, University of Alicante, Sciences Building V, San Vicente del Raspeig Campus, PO Box 99, Alicante 03080, Spain
| | - Dan Tan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - XiaoYun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
2
|
Bosmon T, Abergel C, Claverie JM. 20 years of research on giant viruses. NPJ VIRUSES 2025; 3:9. [PMID: 40295850 PMCID: PMC11814242 DOI: 10.1038/s44298-025-00093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/23/2025] [Indexed: 04/30/2025]
Abstract
Some twenty years ago, the discovery of the first giant virus, Acanthamoeba polyphaga mimivirus (now mimivirus bradfordmassiliense species), paved the way for the discovery of more than 10 new families of protist-infecting DNA viruses with unexpected diversity in virion shape and size, gene content, genome topology and mode of replication. Following their brief description, we examine how the historical concepts of virology have held up in the light of this new knowledge. Although the initial emphasis was on the gigantism of the newly described viruses infecting amoebae, the subsequent discovery of viruses with intermediate virion and genome sizes gradually re-established a continuum between the smallest and largest viruses within the phylum Nucleocytoviricota.
Collapse
Affiliation(s)
- Tressy Bosmon
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale, Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479, IM2B, IOM), 13288, Marseille Cedex 9, France
| | - Chantal Abergel
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale, Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479, IM2B, IOM), 13288, Marseille Cedex 9, France.
| | - Jean-Michel Claverie
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale, Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479, IM2B, IOM), 13288, Marseille Cedex 9, France.
| |
Collapse
|
3
|
Górniak I, Stephens Z, Erramilli SK, Gawda T, Kossiakoff AA, Zimmer J. Structural insights into translocation and tailored synthesis of hyaluronan. Nat Struct Mol Biol 2025; 32:161-171. [PMID: 39322765 PMCID: PMC11750622 DOI: 10.1038/s41594-024-01389-1] [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: 04/04/2024] [Accepted: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Hyaluronan (HA) is an essential component of the vertebrate extracellular matrix. It is a heteropolysaccharide of N-acetylglucosamine (GlcNAc) and glucuronic acid (GlcA) reaching several megadaltons in healthy tissues. HA is synthesized and translocated in a coupled reaction by HA synthase (HAS). Here, structural snapshots of HAS provide insights into HA biosynthesis, from substrate recognition to HA elongation and translocation. We monitor the extension of a GlcNAc primer with GlcA, reveal the coordination of the uridine diphosphate product by a conserved gating loop and capture the opening of a translocation channel to coordinate a translocating HA polymer. Furthermore, we identify channel-lining residues that modulate HA product lengths. Integrating structural and biochemical analyses suggests an avenue for polysaccharide engineering based on finely tuned enzymatic activity and HA coordination.
Collapse
Affiliation(s)
- Ireneusz Górniak
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Zachery Stephens
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Satchal K Erramilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Tomasz Gawda
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Jochen Zimmer
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Wei M, Huang Y, Zhu J, Qiao Y, Xiao N, Jin M, Gao H, Huang Y, Hu X, Li O. Advances in hyaluronic acid production: Biosynthesis and genetic engineering strategies based on Streptococcus - A review. Int J Biol Macromol 2024; 270:132334. [PMID: 38744368 DOI: 10.1016/j.ijbiomac.2024.132334] [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: 11/28/2023] [Revised: 05/02/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Hyaluronic acid (HA), which is a highly versatile glycosaminoglycan, is widely applied across the fields of food, cosmetics, and pharmaceuticals. It is primary produced through Streptococcus fermentation, but the product presents inherent challenges concerning consistency and potential pathogenicity. However, recent strides in molecular biology have paved the way for genetic engineering, which facilitates the creation of high-yield, nonpathogenic strains adept at synthesizing HA with specific molecular weights. This comprehensive review extensively explores the molecular biology underpinning pivotal HA synthase genes, which elucidates the intricate mechanisms governing HA synthesis. Moreover, it delineates various strategies employed in engineering HA-producing strains.
Collapse
Affiliation(s)
- Mengmeng Wei
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Ying Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Junyuan Zhu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Yufan Qiao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Na Xiao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Mengying Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Han Gao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Yitie Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Xiufang Hu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China
| | - Ou Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310000, PR China.
| |
Collapse
|
5
|
Balestreri C, Schroeder DC, Sampedro F, Marqués G, Palowski A, Urriola PE, van de Ligt JLG, Yancy HF, Shurson GC. Unexpected thermal stability of two enveloped megaviruses, Emiliania huxleyi virus and African swine fever virus, as measured by viability PCR. Virol J 2024; 21:1. [PMID: 38172919 PMCID: PMC10765680 DOI: 10.1186/s12985-023-02272-z] [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: 04/20/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The particle structure of Emiliania huxleyi virus (EhV), an algal infecting member of nucleocytoplasmic large DNA viruses (NCLDVs), contains an outer lipid membrane envelope similar to that found in animal viruses such as African swine fever virus (ASFV). Despite both being enveloped NCLDVs, EhV and ASFV are known for their stability outside their host environment. METHOD Here we report for the first time, the application of a viability qPCR (V-qPCR) method to describe the unprecedented and similar virion thermal stability of both EhV and ASFV. This result contradicts the cell culture-based assay method that suggests that virus "infectivity" is lost in a matter of seconds (for EhV) and minutes (for ASFV) at temperature greater than 50 °C. Confocal microscopy and analytical flow cytometry methods was used to validate the V-qPCR data for EhV. RESULTS We observed that both EhV and ASFV particles has unprecedented thermal tolerances. These two NCLDVs are exceptions to the rule that having an enveloped virion anatomy is a predicted weakness, as is often observed in enveloped RNA viruses (i.e., the viruses causing Porcine Reproductive and Respiratory Syndrome (PRRS), COVID-19, Ebola, or seasonal influenza). Using the V-qPCR method, we confirm that no PRRSV particles were detectable after 20 min of exposure to temperatures up to 100 °C. We also show that the EhV particles that remain after 50 °C 20 min exposure was in fact still infectious only after the three blind passages in bioassay experiments. CONCLUSIONS This study raises the possibility that ASFV is not always eliminated or contained after applying time and temperature inactivation treatments in current decontamination or biosecurity protocols. This observation has practical implications for industries involved in animal health and food security. Finally, we propose that EhV could be used as a surrogate for ASFV under certain circumstances.
Collapse
Affiliation(s)
- Cecilia Balestreri
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, 55108, USA
| | - Declan C Schroeder
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, 55108, USA.
| | - Fernando Sampedro
- Environmental Health Sciences Division, University of Minnesota, St. Paul, MN, 55455, USA
| | - Guillermo Marqués
- Department of Neuroscience, University Imaging Centers, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Amanda Palowski
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, 55108, USA
| | - Pedro E Urriola
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, 55108, USA
- Department of Animal Science, University of Minnesota, St. Paul, MN, 55108, USA
| | | | - Haile F Yancy
- U.S. Food and Drug Administration, Center for Veterinary Medicine, Laurel, MD, 20708, USA
| | - Gerald C Shurson
- Department of Animal Science, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
6
|
DeAngelis PL, Zimmer J. Hyaluronan synthases; mechanisms, myths, & mysteries of three types of unique bifunctional glycosyltransferases. Glycobiology 2023; 33:1117-1127. [PMID: 37769351 PMCID: PMC10939387 DOI: 10.1093/glycob/cwad075] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
Hyaluronan (HA), the essential [-3-GlcNAc-1-β-4-GlcA-1-β-]n matrix polysaccharide in vertebrates and molecular camouflage coating in select pathogens, is polymerized by "HA synthase" (HAS) enzymes. The first HAS identified three decades ago opened the window for new insights and biotechnological tools. This review discusses current understanding of HA biosynthesis, its biotechnological utility, and addresses some misconceptions in the literature. HASs are fascinating enzymes that polymerize two different UDP-activated sugars via different glycosidic linkages. Therefore, these catalysts were the first examples to break the "one enzyme/one sugar transferred" dogma. Three distinct types of these bifunctional glycosyltransferases (GTs) with disparate architectures and reaction modes are known. Based on biochemical and structural work, we present an updated classification system. Class I membrane-integrated HASs employ a processive chain elongation mechanism and secrete HA across the plasma membrane. This complex operation is accomplished by functionally integrating a cytosolic catalytic domain with a channel-forming transmembrane region. Class I enzymes, containing a single GT family-2 (GT-2) module that adds both monosaccharide units to the nascent chain, are further subdivided into two groups that construct the polymer with opposite molecular directionalities: Class I-R and I-NR elongate the HA polysaccharide at either the reducing or the non-reducing end, respectively. In contrast, Class II HASs are membrane-associated peripheral synthases with a non-processive, non-reducing end elongation mechanism using two independent GT-2 modules (one for each type of monosaccharide) and require a separate secretion system for HA export. We discuss recent mechanistic insights into HA biosynthesis that promise biotechnological benefits and exciting engineering approaches.
Collapse
Affiliation(s)
- Paul L DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., Oklahoma, OK 73104, United States
| | - Jochen Zimmer
- Department of Molecular Physiology and Biological Physics, Howard Hughes Medical Institute, University of Virginia, 480 Ray C. Hunt Dr, Charlottesville, VA 22908, United States
| |
Collapse
|
7
|
Moniruzzaman M, Erazo Garcia MP, Farzad R, Ha AD, Jivaji A, Karki S, Sheyn U, Stanton J, Minch B, Stephens D, Hancks DC, Rodrigues RAL, Abrahao JS, Vardi A, Aylward FO. Virologs, viral mimicry, and virocell metabolism: the expanding scale of cellular functions encoded in the complex genomes of giant viruses. FEMS Microbiol Rev 2023; 47:fuad053. [PMID: 37740576 PMCID: PMC10583209 DOI: 10.1093/femsre/fuad053] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/29/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023] Open
Abstract
The phylum Nucleocytoviricota includes the largest and most complex viruses known. These "giant viruses" have a long evolutionary history that dates back to the early diversification of eukaryotes, and over time they have evolved elaborate strategies for manipulating the physiology of their hosts during infection. One of the most captivating of these mechanisms involves the use of genes acquired from the host-referred to here as viral homologs or "virologs"-as a means of promoting viral propagation. The best-known examples of these are involved in mimicry, in which viral machinery "imitates" immunomodulatory elements in the vertebrate defense system. But recent findings have highlighted a vast and rapidly expanding array of other virologs that include many genes not typically found in viruses, such as those involved in translation, central carbon metabolism, cytoskeletal structure, nutrient transport, vesicular trafficking, and light harvesting. Unraveling the roles of virologs during infection as well as the evolutionary pathways through which complex functional repertoires are acquired by viruses are important frontiers at the forefront of giant virus research.
Collapse
Affiliation(s)
- Mohammad Moniruzzaman
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Maria Paula Erazo Garcia
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Roxanna Farzad
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Anh D Ha
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Abdeali Jivaji
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Sangita Karki
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Uri Sheyn
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Joshua Stanton
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Benjamin Minch
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Danae Stephens
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Dustin C Hancks
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, United States
| | - Rodrigo A L Rodrigues
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Jonatas S Abrahao
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Assaf Vardi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Frank O Aylward
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
- Center for Emerging, Zoonotic, and Arthropod-Borne Infectious Disease, Virginia Tech, Blacksburg, VA 24061, United States
| |
Collapse
|
8
|
Gilbert NE, LeCleir GR, Pound HL, Strzepek RF, Ellwood MJ, Twining BS, Roux S, Boyd PW, Wilhelm SW. Giant Virus Infection Signatures Are Modulated by Euphotic Zone Depth Strata and Iron Regimes of the Subantarctic Southern Ocean. mSystems 2023; 8:e0126022. [PMID: 36794943 PMCID: PMC10134803 DOI: 10.1128/msystems.01260-22] [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: 12/19/2022] [Accepted: 01/24/2023] [Indexed: 02/17/2023] Open
Abstract
Viruses can alter the abundance, evolution, and metabolism of microorganisms in the ocean, playing a key role in water column biogeochemistry and global carbon cycles. Large efforts to measure the contribution of eukaryotic microorganisms (e.g., protists) to the marine food web have been made, yet the in situ activities of the ecologically relevant viruses that infect these organisms are not well characterized. Viruses within the phylum Nucleocytoviricota ("giant viruses") are known to infect a diverse range of ecologically relevant marine protists, yet how these viruses are influenced by environmental conditions remains under-characterized. By employing metatranscriptomic analyses of in situ microbial communities along a temporal and depth-resolved gradient, we describe the diversity of giant viruses at the Southern Ocean Time Series (SOTS), a site within the subpolar Southern Ocean. Using a phylogeny-guided taxonomic assessment of detected giant virus genomes and metagenome-assembled genomes, we observed depth-dependent structuring of divergent giant virus families mirroring dynamic physicochemical gradients in the stratified euphotic zone. Analyses of transcribed metabolic genes from giant viruses suggest viral metabolic reprogramming of hosts from the surface to a 200-m depth. Lastly, using on-deck incubations reflecting a gradient of iron availability, we show that modulating iron regimes influences the activity of giant viruses in the field. Specifically, we show enhanced infection signatures of giant viruses under both iron-replete and iron-limited conditions. Collectively, these results expand our understanding of how the water column's vertical biogeography and chemical surroundings affect an important group of viruses within the Southern Ocean. IMPORTANCE The biology and ecology of marine microbial eukaryotes is known to be constrained by oceanic conditions. In contrast, how viruses that infect this important group of organisms respond to environmental change is less well known, despite viruses being recognized as key microbial community members. Here, we address this gap in our understanding by characterizing the diversity and activity of "giant" viruses within an important region in the sub-Antarctic Southern Ocean. Giant viruses are double-stranded DNA (dsDNA) viruses of the phylum Nucleocytoviricota and are known to infect a wide range of eukaryotic hosts. By employing a metatranscriptomics approach using both in situ samples and microcosm manipulations, we illuminated both the vertical biogeography and how changing iron availability affects this primarily uncultivated group of protist-infecting viruses. These results serve as a foundation for our understanding of how the open ocean water column structures the viral community, which can be used to guide models of the viral impact on marine and global biogeochemical cycling.
Collapse
Affiliation(s)
- Naomi E. Gilbert
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Gary R. LeCleir
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Helena L. Pound
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| | - Robert F. Strzepek
- Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, Tasmania, Australia
| | - Michael J. Ellwood
- Research School of Earth Sciences, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | - Simon Roux
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Philip W. Boyd
- Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, Tasmania, Australia
| | - Steven W. Wilhelm
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
9
|
Zamboni F, Wong CK, Collins MN. Hyaluronic acid association with bacterial, fungal and viral infections: Can hyaluronic acid be used as an antimicrobial polymer for biomedical and pharmaceutical applications? Bioact Mater 2023; 19:458-473. [PMID: 35574061 PMCID: PMC9079116 DOI: 10.1016/j.bioactmat.2022.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/23/2022] [Indexed: 12/21/2022] Open
Abstract
The relationships between hyaluronic acid (HA) and pathological microorganisms incite new understandings on microbial infection, tissue penetration, disease progression and lastly, potential treatments. These understandings are important for the advancement of next generation antimicrobial therapeutical strategies for the control of healthcare-associated infections. Herein, this review will focus on the interplay between HA, bacteria, fungi, and viruses. This review will also comprehensively detail and discuss the antimicrobial activity displayed by various HA molecular weights for a variety of biomedical and pharmaceutical applications, including microbiology, pharmaceutics, and tissue engineering.
Collapse
Affiliation(s)
- Fernanda Zamboni
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Ireland
- Health Research Institute, University of Limerick, Ireland
| |
Collapse
|
10
|
Giant Viruses as a Source of Novel Enzymes for Biotechnological Application. Pathogens 2022; 11:pathogens11121453. [PMID: 36558786 PMCID: PMC9787589 DOI: 10.3390/pathogens11121453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The global demand for industrial enzymes has been increasing in recent years, and the search for new sources of these biological products is intense, especially in microorganisms. Most known viruses have limited genetic machinery and, thus, have been overlooked by the enzyme industry for years. However, a peculiar group of viruses breaks this paradigm. Giant viruses of the phylum Nucleocytoviricota infect protists (i.e., algae and amoebae) and have complex genomes, reaching up to 2.7 Mb in length and encoding hundreds of genes. Different giant viruses have robust metabolic machinery, especially those in the Phycodnaviridae and Mimiviridae families. In this review, we present some peculiarities of giant viruses that infect protists and discuss why they should be seen as an outstanding source of new enzymes. We revisited the genomes of representatives of different groups of giant viruses and put together information about their enzymatic machinery, highlighting several genes to be explored in biotechnology involved in carbohydrate metabolism, DNA replication, and RNA processing, among others. Finally, we present additional evidence based on structural biology using chitinase as a model to reinforce the role of giant viruses as a source of novel enzymes for biotechnological application.
Collapse
|
11
|
Shikina E, Kovalevsky R, Shirkovskaya A, Toukach P. Prospective bacterial and fungal sources of hyaluronic acid: A review. Comput Struct Biotechnol J 2022; 20:6214-6236. [PMID: 36420162 PMCID: PMC9676211 DOI: 10.1016/j.csbj.2022.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/05/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
The unique biological and rheological properties make hyaluronic acid a sought-after material for medicine and cosmetology. Due to very high purity requirements for hyaluronic acid in medical applications, the profitability of streptococcal fermentation is reduced. Production of hyaluronic acid by recombinant systems is considered a promising alternative. Variations in combinations of expressed genes and fermentation conditions alter the yield and molecular weight of produced hyaluronic acid. This review is devoted to the current state of hyaluronic acid production by recombinant bacterial and fungal organisms.
Collapse
|
12
|
Speciale I, Notaro A, Abergel C, Lanzetta R, Lowary TL, Molinaro A, Tonetti M, Van Etten JL, De Castro C. The Astounding World of Glycans from Giant Viruses. Chem Rev 2022; 122:15717-15766. [PMID: 35820164 PMCID: PMC9614988 DOI: 10.1021/acs.chemrev.2c00118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Indexed: 12/12/2022]
Abstract
Viruses are a heterogeneous ensemble of entities, all sharing the need for a suitable host to replicate. They are extremely diverse, varying in morphology, size, nature, and complexity of their genomic content. Typically, viruses use host-encoded glycosyltransferases and glycosidases to add and remove sugar residues from their glycoproteins. Thus, the structure of the glycans on the viral proteins have, to date, typically been considered to mimick those of the host. However, the more recently discovered large and giant viruses differ from this paradigm. At least some of these viruses code for an (almost) autonomous glycosylation pathway. These viral genes include those that encode the production of activated sugars, glycosyltransferases, and other enzymes able to manipulate sugars at various levels. This review focuses on large and giant viruses that produce carbohydrate-processing enzymes. A brief description of those harboring these features at the genomic level will be discussed, followed by the achievements reached with regard to the elucidation of the glycan structures, the activity of the proteins able to manipulate sugars, and the organic synthesis of some of these virus-encoded glycans. During this progression, we will also comment on many of the challenging questions on this subject that remain to be addressed.
Collapse
Affiliation(s)
- Immacolata Speciale
- Department
of Agricultural Sciences, University of
Napoli, Via Università
100, 80055 Portici, Italy
| | - Anna Notaro
- Department
of Agricultural Sciences, University of
Napoli, Via Università
100, 80055 Portici, Italy
- Centre
National de la Recherche Scientifique, Information Génomique
& Structurale, Aix-Marseille University, Unité Mixte de Recherche
7256, IMM, IM2B, 13288 Marseille, Cedex 9, France
| | - Chantal Abergel
- Centre
National de la Recherche Scientifique, Information Génomique
& Structurale, Aix-Marseille University, Unité Mixte de Recherche
7256, IMM, IM2B, 13288 Marseille, Cedex 9, France
| | - Rosa Lanzetta
- Department
of Chemical Sciences, University of Napoli, Via Cintia 4, 80126 Napoli, Italy
| | - Todd L. Lowary
- Institute
of Biological Chemistry, Academia Sinica, Academia Road, Section 2, Nangang 11529, Taipei, Taiwan
| | - Antonio Molinaro
- Department
of Chemical Sciences, University of Napoli, Via Cintia 4, 80126 Napoli, Italy
| | - Michela Tonetti
- Department
of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova, 16132 Genova, Italy
| | - James L. Van Etten
- Nebraska
Center for Virology, University of Nebraska, Lincoln, Nebraska 68583-0900, United States
- Department
of Plant Pathology, University of Nebraska, Lincoln, Nebraska 68583-0722, United States
| | - Cristina De Castro
- Department
of Agricultural Sciences, University of
Napoli, Via Università
100, 80055 Portici, Italy
| |
Collapse
|
13
|
Aylward FO, Moniruzzaman M. Viral Complexity. Biomolecules 2022; 12:1061. [PMID: 36008955 PMCID: PMC9405923 DOI: 10.3390/biom12081061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/18/2022] Open
Abstract
Although traditionally viewed as streamlined and simple, discoveries over the last century have revealed that viruses can exhibit surprisingly complex physical structures, genomic organization, ecological interactions, and evolutionary histories. Viruses can have physical dimensions and genome lengths that exceed many cellular lineages, and their infection strategies can involve a remarkable level of physiological remodeling of their host cells. Virus-virus communication and widespread forms of hyperparasitism have been shown to be common in the virosphere, demonstrating that dynamic ecological interactions often shape their success. And the evolutionary histories of viruses are often fraught with complexities, with chimeric genomes including genes derived from numerous distinct sources or evolved de novo. Here we will discuss many aspects of this viral complexity, with particular emphasis on large DNA viruses, and provide an outlook for future research.
Collapse
Affiliation(s)
- Frank O. Aylward
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mohammad Moniruzzaman
- Rosenstiel School of Marine and Atmospheric Science, University of Miami, Coral Gables, FL 33149, USA;
| |
Collapse
|
14
|
Schulz F, Abergel C, Woyke T. Giant virus biology and diversity in the era of genome-resolved metagenomics. Nat Rev Microbiol 2022; 20:721-736. [PMID: 35902763 DOI: 10.1038/s41579-022-00754-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 11/09/2022]
Abstract
The discovery of giant viruses, with capsids as large as some bacteria, megabase-range genomes and a variety of traits typically found only in cellular organisms, was one of the most remarkable breakthroughs in biology. Until recently, most of our knowledge of giant viruses came from ~100 species-level isolates for which genome sequences were available. However, these isolates were primarily derived from laboratory-based co-cultivation with few cultured protists and algae and, thus, did not reflect the true diversity of giant viruses. Although virus co-cultures enabled valuable insights into giant virus biology, many questions regarding their origin, evolution and ecological importance remain unanswered. With advances in sequencing technologies and bioinformatics, our understanding of giant viruses has drastically expanded. In this Review, we summarize our understanding of giant virus diversity and biology based on viral isolates as laboratory cultivation has enabled extensive insights into viral morphology and infection strategies. We then explore how cultivation-independent approaches have heightened our understanding of the coding potential and diversity of the Nucleocytoviricota. We discuss how metagenomics has revolutionized our perspective of giant viruses by revealing their distribution across our planet's biomes, where they impact the biology and ecology of a wide range of eukaryotic hosts and ultimately affect global nutrient cycles.
Collapse
Affiliation(s)
- Frederik Schulz
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Chantal Abergel
- Aix Marseille University, CNRS, IGS UMR7256, IMM FR3479, IM2B, IO, Marseille, France
| | - Tanja Woyke
- DOE Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA. .,University of California Merced, Merced, CA, USA.
| |
Collapse
|
15
|
Maloney FP, Kuklewicz J, Corey RA, Bi Y, Ho R, Mateusiak L, Pardon E, Steyaert J, Stansfeld PJ, Zimmer J. Structure, substrate recognition and initiation of hyaluronan synthase. Nature 2022; 604:195-201. [PMID: 35355017 PMCID: PMC9358715 DOI: 10.1038/s41586-022-04534-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 02/08/2022] [Indexed: 11/09/2022]
Abstract
Hyaluronan is an acidic heteropolysaccharide comprising alternating N-acetylglucosamine and glucuronic acid sugars that is ubiquitously expressed in the vertebrate extracellular matrix1. The high-molecular-mass polymer modulates essential physiological processes in health and disease, including cell differentiation, tissue homeostasis and angiogenesis2. Hyaluronan is synthesized by a membrane-embedded processive glycosyltransferase, hyaluronan synthase (HAS), which catalyses the synthesis and membrane translocation of hyaluronan from uridine diphosphate-activated precursors3,4. Here we describe five cryo-electron microscopy structures of a viral HAS homologue at different states during substrate binding and initiation of polymer synthesis. Combined with biochemical analyses and molecular dynamics simulations, our data reveal how HAS selects its substrates, hydrolyses the first substrate to prime the synthesis reaction, opens a hyaluronan-conducting transmembrane channel, ensures alternating substrate polymerization and coordinates hyaluronan inside its transmembrane pore. Our research suggests a detailed model for the formation of an acidic extracellular heteropolysaccharide and provides insights into the biosynthesis of one of the most abundant and essential glycosaminoglycans in the human body.
Collapse
Affiliation(s)
- Finn P Maloney
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jeremi Kuklewicz
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Robin A Corey
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Yunchen Bi
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Ruoya Ho
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lukasz Mateusiak
- Laboratory for In Vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, VUB, Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, VUB, Brussels, Belgium
| | - Phillip J Stansfeld
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK
| | - Jochen Zimmer
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
16
|
Ma Y, Qiu Y, Yu C, Li S, Xu H. Design and construction of a Bacillus amyloliquefaciens cell factory for hyaluronic acid synthesis from Jerusalem artichoke inulin. Int J Biol Macromol 2022; 205:410-418. [PMID: 35202630 DOI: 10.1016/j.ijbiomac.2022.02.100] [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: 09/26/2021] [Revised: 01/21/2022] [Accepted: 02/16/2022] [Indexed: 11/05/2022]
Abstract
Hyaluronic acid (HA), a high-value biomacromolecule, has wide applications in medical, cosmetic and food fields. Currently, employing the safe-grade microorganisms for de novo biosynthesis of HA from renewable substrates has become a promising alternative. In this study, we established a Bacillus amyloliquefaciens strain as platform for HA production from Jerusalem artichoke inulin. Firstly, the different HA and UDP-GlcUA synthase genes were introduced into B. amyloliquefaciens to construct the HA synthesis pathway. Secondly, the byproduct polysaccharides were removed by knocking sacB and epsA-O using CRISPR/Cas9n system, resulting in a 13% increase in HA production. Finally, 2.89 g/L HA with a high molecular weight of 1.5 MDa was obtained after optimizing fermentation conditions and adding osmotic agents. This study demonstrates the engineered B. amyloliquefaciens can effectively synthesize HA with Jerusalem artichoke inulin and provides a green route for HA production.
Collapse
Affiliation(s)
- Yanqin Ma
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China
| | - Yibin Qiu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China; College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, PR China; State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China
| | - Caiyuan Yu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China
| | - Sha Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China.
| | - Hong Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China.
| |
Collapse
|
17
|
Qiu Y, Ma Y, Huang Y, Li S, Xu H, Su E. Current advances in the biosynthesis of hyaluronic acid with variable molecular weights. Carbohydr Polym 2021; 269:118320. [PMID: 34294332 DOI: 10.1016/j.carbpol.2021.118320] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/05/2021] [Accepted: 06/06/2021] [Indexed: 12/26/2022]
Abstract
Hyaluronic acid (HA) is a naturally formed acidic mucopolysaccharide, with excellent moisturising properties and used widely in the medicine, cosmetics, and food industries. The industrial production of specific molecular weight HA has become imperative. Different biological activities and physiological functions of HA mainly depend on the degree of polymerisation. This article reviews the research status and development prospects of the green biosynthesis and molecular weight regulation of HA. There is an application-based prerequisite of specific molecular weight of HA that could be regulated either during the fermentation process or via a controlled HA degradation process. This work provides an important theoretical basis for the downstream efficient production of diversified HA, which will further accelerate the research applications of HA and provide a good scientific basis and method reference for the study of the molecular weight regulation of similar biopolymers.
Collapse
Affiliation(s)
- Yibin Qiu
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, PR China; State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China; Yangzhou Rixing Bio-Tech Co., Ltd., Yangzhou 225601, PR China.
| | - Yanqin Ma
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China
| | - Yanyan Huang
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Sha Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China
| | - Hong Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, PR China
| | - Erzheng Su
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, PR China.
| |
Collapse
|
18
|
Novel Capsular Polysaccharide from Lobochlamys segnis. POLYSACCHARIDES 2021. [DOI: 10.3390/polysaccharides2010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In recent years there has been a significant effort from food, nutraceutical, cosmeceutical, pharmaceutical, and biomedical industries to discover and develop new natural ingredients. Microalgae have been recognised as potential sources of high value chemicals, with most attention focused upon antioxidants, pigments, and specialty oils. An under-exploited group of biochemicals produced by microalgae are extracellular polymeric substances (EPS). Lobochlamys segnis (formerly called Chlamydomonas segnis) was previously reported to produce a large amount of capsular polysaccharide (CPS) closely related to hyaluronan (HA) under stress conditions. In this work, the purified CPS was characterised and shown to have an average molecular mass (Mn) of about 3.7 MDa, and displayed a highly branched random coil structure unlike the simple repeating linear HA polysaccharide. Chemical analysis showed the presence of galactose, glucuronic acid, and glucose sugars confirming that the product has a different composition to that of HA. Mixotrophic growth and stress conditions were identified and improved upon with a pH control system using acetic acid solution to induce efficient production of CPS. Extraction and purification conditions were also identified exploiting the high Mn of the product. The CPS showed thickening properties and both significant antioxidant capacity and reducing power, which could have commercial applications. This is the first report on the characterization of this novel polysaccharide with non-Newtonian properties similar to HA.
Collapse
|
19
|
Mohd Nadzir M, Nurhayati RW, Idris FN, Nguyen MH. Biomedical Applications of Bacterial Exopolysaccharides: A Review. Polymers (Basel) 2021; 13:530. [PMID: 33578978 PMCID: PMC7916691 DOI: 10.3390/polym13040530] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Bacterial exopolysaccharides (EPSs) are an essential group of compounds secreted by bacteria. These versatile EPSs are utilized individually or in combination with different materials for a broad range of biomedical field functions. The various applications can be explained by the vast number of derivatives with useful properties that can be controlled. This review offers insight on the current research trend of nine commonly used EPSs, their biosynthesis pathways, their characteristics, and the biomedical applications of these relevant bioproducts.
Collapse
Affiliation(s)
- Masrina Mohd Nadzir
- School of Chemical Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal 14300, Malaysia;
| | - Retno Wahyu Nurhayati
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia;
- Stem Cell and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No. 6, Jakarta 10430, Indonesia
| | - Farhana Nazira Idris
- School of Chemical Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal 14300, Malaysia;
| | - Minh Hong Nguyen
- Faculty of Biotechnology, Chemistry and Environmental Engineering, Phenikaa University, Hanoi 12116, Vietnam;
- Bioresource Research Center, Phenikaa University, Hanoi 12116, Vietnam
| |
Collapse
|
20
|
Sun TW, Yang CL, Kao TT, Wang TH, Lai MW, Ku C. Host Range and Coding Potential of Eukaryotic Giant Viruses. Viruses 2020; 12:E1337. [PMID: 33233432 PMCID: PMC7700475 DOI: 10.3390/v12111337] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Giant viruses are a group of eukaryotic double-stranded DNA viruses with large virion and genome size that challenged the traditional view of virus. Newly isolated strains and sequenced genomes in the last two decades have substantially advanced our knowledge of their host diversity, gene functions, and evolutionary history. Giant viruses are now known to infect hosts from all major supergroups in the eukaryotic tree of life, which predominantly comprises microbial organisms. The seven well-recognized viral clades (taxonomic families) have drastically different host range. Mimiviridae and Phycodnaviridae, both with notable intrafamilial genome variation and high abundance in environmental samples, have members that infect the most diverse eukaryotic lineages. Laboratory experiments and comparative genomics have shed light on the unprecedented functional potential of giant viruses, encoding proteins for genetic information flow, energy metabolism, synthesis of biomolecules, membrane transport, and sensing that allow for sophisticated control of intracellular conditions and cell-environment interactions. Evolutionary genomics can illuminate how current and past hosts shape viral gene repertoires, although it becomes more obscure with divergent sequences and deep phylogenies. Continued works to characterize giant viruses from marine and other environments will further contribute to our understanding of their host range, coding potential, and virus-host coevolution.
Collapse
Affiliation(s)
- Tsu-Wang Sun
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
| | - Chia-Ling Yang
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
| | - Tzu-Tong Kao
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
| | - Tzu-Haw Wang
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
| | - Ming-Wei Lai
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
| | - Chuan Ku
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan; (T.-W.S.); (C.-L.Y.); (T.-T.K.); (T.-H.W.); (M.-W.L.)
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
| |
Collapse
|
21
|
Speciale I, Laugieri ME, Noel E, Lin S, Lowary TL, Molinaro A, Duncan GA, Agarkova IV, Garozzo D, Tonetti MG, Van Etten JL, De Castro C. Chlorovirus PBCV-1 protein A064R has three of the transferase activities necessary to synthesize its capsid protein N-linked glycans. Proc Natl Acad Sci U S A 2020; 117:28735-28742. [PMID: 33139538 PMCID: PMC7682578 DOI: 10.1073/pnas.2016626117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Paramecium bursaria chlorella virus-1 (PBCV-1) is a large double-stranded DNA (dsDNA) virus that infects the unicellular green alga Chlorella variabilis NC64A. Unlike many other viruses, PBCV-1 encodes most, if not all, of the enzymes involved in the synthesis of the glycans attached to its major capsid protein. Importantly, these glycans differ from those reported from the three domains of life in terms of structure and asparagine location in the sequon of the protein. Previous data collected from 20 PBCV-1 spontaneous mutants (or antigenic variants) suggested that the a064r gene encodes a glycosyltransferase (GT) with three domains, each with a different function. Here, we demonstrate that: domain 1 is a β-l-rhamnosyltransferase; domain 2 is an α-l-rhamnosyltransferase resembling only bacterial proteins of unknown function, and domain 3 is a methyltransferase that methylates the C-2 hydroxyl group of the terminal α-l-rhamnose (Rha) unit. We also establish that methylation of the C-3 hydroxyl group of the terminal α-l-Rha is achieved by another virus-encoded protein A061L, which requires an O-2 methylated substrate. This study, thus, identifies two of the glycosyltransferase activities involved in the synthesis of the N-glycan of the viral major capsid protein in PBCV-1 and establishes that a single protein A064R possesses the three activities needed to synthetize the 2-OMe-α-l-Rha-(1→2)-β-l-Rha fragment. Remarkably, this fragment can be attached to any xylose unit.
Collapse
Affiliation(s)
- Immacolata Speciale
- Department of Chemical Sciences, University of Napoli Federico II, 80126 Napoli, Italy
- Department of Agricultural Sciences, University of Napoli Federico II, 80055 Portici NA, Italy
| | - Maria Elena Laugieri
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova, 16132 Genova, Italy
| | - Eric Noel
- Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900
- School of Biological Sciences, University of Nebraska, Lincoln, NE 68588-0118
| | - Sicheng Lin
- Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Centre, Edmonton, AB T6G 2G2 , Canada
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Centre, Edmonton, AB T6G 2G2 , Canada
- Institute of Biological Chemistry, Academia Sinica, Nangang, 11529 Taipei, Taiwan
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, 80126 Napoli, Italy
| | - Garry A Duncan
- Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900
| | - Irina V Agarkova
- Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900
- Department of Plant Pathology, University of Nebraska, Lincoln, NE 68583-0722
| | - Domenico Garozzo
- CNR, Institute for Polymers, Composites and Biomaterials, 95126 Catania, Italy
| | - Michela G Tonetti
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova, 16132 Genova, Italy;
| | - James L Van Etten
- Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900;
- Department of Plant Pathology, University of Nebraska, Lincoln, NE 68583-0722
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Napoli Federico II, 80055 Portici NA, Italy;
| |
Collapse
|
22
|
Van Etten JL, Agarkova IV, Dunigan DD. Chloroviruses. Viruses 2019; 12:E20. [PMID: 31878033 PMCID: PMC7019647 DOI: 10.3390/v12010020] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 12/20/2022] Open
Abstract
Chloroviruses are large dsDNA, plaque-forming viruses that infect certain chlorella-like green algae; the algae are normally mutualistic endosymbionts of protists and metazoans and are often referred to as zoochlorellae. The viruses are ubiquitous in inland aqueous environments throughout the world and occasionally single types reach titers of thousands of plaque-forming units per ml of native water. The viruses are icosahedral in shape with a spike structure located at one of the vertices. They contain an internal membrane that is required for infectivity. The viral genomes are 290 to 370 kb in size, which encode up to 16 tRNAs and 330 to ~415 proteins, including many not previously seen in viruses. Examples include genes encoding DNA restriction and modification enzymes, hyaluronan and chitin biosynthetic enzymes, polyamine biosynthetic enzymes, ion channel and transport proteins, and enzymes involved in the glycan synthesis of the virus major capsid glycoproteins. The proteins encoded by many of these viruses are often the smallest or among the smallest proteins of their class. Consequently, some of the viral proteins are the subject of intensive biochemical and structural investigation.
Collapse
Affiliation(s)
- James L. Van Etten
- Department of Plant Pathology, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583-0900, USA; (I.V.A.); (D.D.D.)
| | | | | |
Collapse
|
23
|
Schulte S, Doss SS, Jeeva P, Ananth M, Blank LM, Jayaraman G. Exploiting the diversity of streptococcal hyaluronan synthases for the production of molecular weight–tailored hyaluronan. Appl Microbiol Biotechnol 2019; 103:7567-7581. [DOI: 10.1007/s00253-019-10023-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 11/28/2022]
|
24
|
Speciale I, Duncan GA, Unione L, Agarkova IV, Garozzo D, Jimenez-Barbero J, Lin S, Lowary TL, Molinaro A, Noel E, Laugieri ME, Tonetti MG, Van Etten JL, De Castro C. The N-glycan structures of the antigenic variants of chlorovirus PBCV-1 major capsid protein help to identify the virus-encoded glycosyltransferases. J Biol Chem 2019; 294:5688-5699. [PMID: 30737276 DOI: 10.1074/jbc.ra118.007182] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/07/2019] [Indexed: 11/06/2022] Open
Abstract
The chlorovirus Paramecium bursaria chlorella virus 1 (PBCV-1) is a large dsDNA virus that infects the microalga Chlorella variabilis NC64A. Unlike most other viruses, PBCV-1 encodes most, if not all, of the machinery required to glycosylate its major capsid protein (MCP). The structures of the four N-linked glycans from the PBCV-1 MCP consist of nonasaccharides, and similar glycans are not found elsewhere in the three domains of life. Here, we identified the roles of three virus-encoded glycosyltransferases (GTs) that have four distinct GT activities in glycan synthesis. Two of the three GTs were previously annotated as GTs, but the third GT was identified in this study. We determined the GT functions by comparing the WT glycan structures from PBCV-1 with those from a set of PBCV-1 spontaneous GT gene mutants resulting in antigenic variants having truncated glycan structures. According to our working model, the virus gene a064r encodes a GT with three domains: domain 1 has a β-l-rhamnosyltransferase activity, domain 2 has an α-l-rhamnosyltransferase activity, and domain 3 is a methyltransferase that decorates two positions in the terminal α-l-rhamnose (Rha) unit. The a075l gene encodes a β-xylosyltransferase that attaches the distal d-xylose (Xyl) unit to the l-fucose (Fuc) that is part of the conserved N-glycan core region. Last, gene a071r encodes a GT that is involved in the attachment of a semiconserved element, α-d-Rha, to the same l-Fuc in the core region. Our results uncover GT activities that assemble four of the nine residues of the PBCV-1 MCP N-glycans.
Collapse
Affiliation(s)
- Immacolata Speciale
- From the Department of Agricultural Sciences, University of Napoli Federico II, Via Università 100, 80055 Portici NA, Italy
| | - Garry A Duncan
- the Department of Biology, Nebraska Wesleyan University, Lincoln, Nebraska 68504-2794
| | - Luca Unione
- the Chemical Glycobiology Lab, CIC bioGUNE, Bizkaia Technology Park, Bld 800, 48170 Derio, Spain
| | - Irina V Agarkova
- the Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska 68583-0900.,the Department of Plant Pathology, University of Nebraska, Lincoln, Nebraska 68583-0722
| | - Domenico Garozzo
- Institute for Polymers, Composites, and Biomaterials, CNR, Via P. Gaifami 18, 95126 Catania, Italy
| | - Jesus Jimenez-Barbero
- the Chemical Glycobiology Lab, CIC bioGUNE, Bizkaia Technology Park, Bld 800, 48170 Derio, Spain.,the Basque Foundation for Science (IKERBASQUE), 48940 Bilbao, Spain.,the Department of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country, EHU-UPV, 48940 Leioa, Spain
| | - Sicheng Lin
- the Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Centre, Edmonton, Alberta T6G 2G2, Canada
| | - Todd L Lowary
- the Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux Chemistry Centre, Edmonton, Alberta T6G 2G2, Canada
| | - Antonio Molinaro
- the Department of Chemical Sciences, Università of Napoli Federico II, 80126 Napoli, Italy
| | - Eric Noel
- the Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska 68583-0900.,the School of Biological Sciences, University of Nebraska, Lincoln, Nebraska 68588-0118, and
| | - Maria Elena Laugieri
- the Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova, Viale Benedetto XV/1, 16132 Genova, Italy
| | - Michela G Tonetti
- the Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova, Viale Benedetto XV/1, 16132 Genova, Italy
| | - James L Van Etten
- the Nebraska Center for Virology, University of Nebraska, Lincoln, Nebraska 68583-0900, .,the Department of Plant Pathology, University of Nebraska, Lincoln, Nebraska 68583-0722
| | - Cristina De Castro
- From the Department of Agricultural Sciences, University of Napoli Federico II, Via Università 100, 80055 Portici NA, Italy,
| |
Collapse
|
25
|
Blackburn MR, Hubbard C, Kiessling V, Bi Y, Kloss B, Tamm LK, Zimmer J. Distinct reaction mechanisms for hyaluronan biosynthesis in different kingdoms of life. Glycobiology 2018; 28:108-121. [PMID: 29190396 PMCID: PMC6192386 DOI: 10.1093/glycob/cwx096] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022] Open
Abstract
Hyaluronan (HA) is an acidic high molecular weight cell surface polysaccharide ubiquitously expressed by vertebrates, some pathogenic bacteria and even viruses. HA modulates many essential physiological processes and is implicated in numerous pathological conditions ranging from autoimmune diseases to cancer. In various pathogens, HA functions as a non-immunogenic surface polymer that reduces host immune responses. It is a linear polymer of strictly alternating glucuronic acid and N-acetylglucosamine units synthesized by HA synthase (HAS), a membrane-embedded family-2 glycosyltransferase. The enzyme synthesizes HA and secretes the polymer through a channel formed by its own membrane-integrated domain. To reveal how HAS achieves these tasks, we determined the biologically functional units of bacterial and viral HAS in a lipid bilayer environment by co-immunoprecipitation, single molecule fluorescence photobleaching, and site-specific cross-linking analyses. Our results demonstrate that bacterial HAS functions as an obligate homo-dimer with two functional HAS copies required for catalytic activity. In contrast, the viral enzyme, closely related to vertebrate HAS, functions as a monomer. Using site-specific cross-linking, we identify the dimer interface of bacterial HAS and show that the enzyme uses a reaction mechanism distinct from viral HAS that necessitates a dimeric assembly.
Collapse
Affiliation(s)
- Matthew R Blackburn
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| | - Caitlin Hubbard
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| | - Volker Kiessling
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yunchen Bi
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| | - Brian Kloss
- Center on Membrane Protein Production and Analysis (COMPPÅ), New York Structural Biology Center (NYSBC), 89 Convent Avenue, New York, NY 10027, USA
| | - Lukas K Tamm
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jochen Zimmer
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, USA
| |
Collapse
|
26
|
Green DE, DeAngelis PL. Identification of a chondroitin synthase from an unexpected source, the green sulfur bacterium Chlorobium phaeobacteroides. Glycobiology 2017; 27:469-476. [PMID: 28104786 PMCID: PMC5444263 DOI: 10.1093/glycob/cwx008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 11/14/2022] Open
Abstract
Glycosaminoglycans (GAGs) are known to be present in all animals as well as some pathogenic microbes. Chondroitin sulfate is the most abundant GAG in mammals where it has various structural and adhesion roles. The Gram-negative bacteria Pasteurella multocida Type F and Escherichia coli K4 produce extracellular capsules composed of unsulfated chondroitin or a fructosylated chondroitin, respectively. Such polysaccharides that are structurally related to host molecules do not generally provoke a strong antibody response thus are thought to be employed as molecular camouflage during infection. We observed a sequence from the photosynthetic green sulfur bacteria, Chlorobium phaeobacteroides DSM 266, which was very similar (~62% identical) to the open reading frames of the known bifunctional chondroitin synthases (PmCS and KfoC); some segments are strikingly conserved amongst the three proteins. Recombinant E. coli-derived Chlorobium enzyme preparations were found to possess bona fide chondroitin synthase activity in vitro. This new catalyst, CpCS, however, has a more promiscuous acceptor usage than the prototypical PmCS, which may be of utility in novel chimeric GAG syntheses. The finding of such a similar chondroitin synthase enzyme in C. phaeobacteroides is unexpected for several reasons including (a) a free-living nonpathogenic organism should not "need" an animal self molecule for protection, (b) the Proteobacteria and the green sulfur bacterial lineages diverged ~2.5-3 billion years ago and (c) the ecological niches of these bacteria are not thought to overlap substantially to facilitate horizontal gene transfer. CpCS provides insight into the structure/function relationship of this class of enzymes.
Collapse
Affiliation(s)
- Dixy E Green
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., BMSB 853,Oklahoma City, OK73126-0901, USA
| | - Paul L DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., BMSB 853,Oklahoma City, OK73126-0901, USA
| |
Collapse
|
27
|
Van Etten JL, Agarkova I, Dunigan DD, Tonetti M, De Castro C, Duncan GA. Chloroviruses Have a Sweet Tooth. Viruses 2017; 9:E88. [PMID: 28441734 PMCID: PMC5408694 DOI: 10.3390/v9040088] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 01/16/2023] Open
Abstract
Chloroviruses are large double-stranded DNA (dsDNA) viruses that infect certain isolates of chlorella-like green algae. They contain up to approximately 400 protein-encoding genes and 16 transfer RNA (tRNA) genes. This review summarizes the unexpected finding that many of the chlorovirus genes encode proteins involved in manipulating carbohydrates. These include enzymes involved in making extracellular polysaccharides, such as hyaluronan and chitin, enzymes that make nucleotide sugars, such as GDP-L-fucose and GDP-D-rhamnose and enzymes involved in the synthesis of glycans attached to the virus major capsid proteins. This latter process differs from that of all other glycoprotein containing viruses that traditionally use the host endoplasmic reticulum and Golgi machinery to synthesize and transfer the glycans.
Collapse
Affiliation(s)
- James L Van Etten
- Department of Plant Pathology and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583-0900, USA.
| | - Irina Agarkova
- Department of Plant Pathology and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583-0900, USA.
| | - David D Dunigan
- Department of Plant Pathology and Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583-0900, USA.
| | - Michela Tonetti
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genova Viale Benedetto XV/1, 16132 Genova, Italy.
| | - Christina De Castro
- Department of Agricultural Sciences, University of Napoli, Via Università 100, 80055 Portici, NA, Italy.
| | - Garry A Duncan
- Department of Biology, Nebraska Wesleyan University, Lincoln, NE 68504-2796, USA.
| |
Collapse
|
28
|
Parmar KM, Gaikwad SL, Dhakephalkar PK, Kothari R, Singh RP. Intriguing Interaction of Bacteriophage-Host Association: An Understanding in the Era of Omics. Front Microbiol 2017; 8:559. [PMID: 28439260 PMCID: PMC5383658 DOI: 10.3389/fmicb.2017.00559] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/16/2017] [Indexed: 01/09/2023] Open
Abstract
Innovations in next-generation sequencing technology have introduced new avenues in microbial studies through “omics” approaches. This technology has considerably augmented the knowledge of the microbial world without isolation prior to their identification. With an enormous volume of bacterial “omics” data, considerable attempts have been recently invested to improve an insight into virosphere. The interplay between bacteriophages and their host has created a significant influence on the biogeochemical cycles, microbial diversity, and bacterial population regulation. This review highlights various concepts such as genomics, transcriptomics, proteomics, and metabolomics to infer the phylogenetic affiliation and function of bacteriophages and their impact on diverse microbial communities. Omics technologies illuminate the role of bacteriophage in an environment, the influences of phage proteins on the bacterial host and provide information about the genes important for interaction with bacteria. These investigations will reveal some of bio-molecules and biomarkers of the novel phage which demand to be unveiled.
Collapse
Affiliation(s)
| | | | | | - Ramesh Kothari
- Department of Biosciences, Saurashtra UniversityRajkot, India
| | | |
Collapse
|
29
|
Tsepilov RN, Beloded AV. Hyaluronic Acid--an "Old" Molecule with "New" Functions: Biosynthesis and Depolymerization of Hyaluronic Acid in Bacteria and Vertebrate Tissues Including during Carcinogenesis. BIOCHEMISTRY (MOSCOW) 2016; 80:1093-108. [PMID: 26555463 DOI: 10.1134/s0006297915090011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hyaluronic acid is an evolutionarily ancient molecule commonly found in vertebrate tissues and capsules of some bacteria. Here we review modern data regarding structure, properties, and biological functions of hyaluronic acid in mammals and Streptococcus spp. bacteria. Various aspects of biogenesis and degradation of hyaluronic acid are discussed, biosynthesis and degradation metabolic pathways for glycosaminoglycan together with involved enzymes are described, and vertebrate and bacterial hyaluronan synthase genes are characterized. Special attention is given to the mechanisms underlying the biological action of hyaluronic acid as well as the interaction between polysaccharide and various proteins. In addition, all known signaling pathways involving hyaluronic acid are outlined. Impaired hyaluronic acid metabolism, changes in biopolymer molecular weight, hyaluronidase activity, and enzyme isoforms often accompany carcinogenesis. The interaction between cells and hyaluronic acid from extracellular matrix that may be important during malignant change is discussed. An expected role for high molecular weight hyaluronic acid in resistance of naked mole rat to oncologic diseases and the protective role of hyaluronic acid in bacteria are discussed.
Collapse
Affiliation(s)
- R N Tsepilov
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian Academy of Medical Sciences, Moscow, 123098, Russia.
| | | |
Collapse
|
30
|
Zhao X, Chen Z, Gu G, Guo Z. Recent advances in the research of bacterial glucuronosyltransferases. J Carbohydr Chem 2016. [DOI: 10.1080/07328303.2016.1205597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
31
|
Virocell Metabolism: Metabolic Innovations During Host-Virus Interactions in the Ocean. Trends Microbiol 2016; 24:821-832. [PMID: 27395772 DOI: 10.1016/j.tim.2016.06.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/06/2016] [Accepted: 06/13/2016] [Indexed: 11/24/2022]
Abstract
Marine viruses are considered to be major ecological, evolutionary, and biogeochemical drivers of the marine environment, responsible for nutrient recycling and determining species composition. Viruses can re-shape their host's metabolic network during infection, generating the virocell-a unique metabolic state that supports their specific requirement. Here we discuss the concept of 'virocell metabolism' and its formation by rewiring of host-encoded metabolic networks, or by introducing virus-encoded auxiliary metabolic genes which provide the virocell with novel metabolic capabilities. The ecological role of marine viruses is commonly assessed by their relative abundance and phylogenetic diversity, lacking the ability to assess the dynamics of active viral infection. The new ability to define a unique metabolic state of the virocell will expand the current virion-centric approaches in order to quantify the impact of marine viruses on microbial food webs.
Collapse
|
32
|
de Oliveira JD, Carvalho LS, Gomes AMV, Queiroz LR, Magalhães BS, Parachin NS. Genetic basis for hyper production of hyaluronic acid in natural and engineered microorganisms. Microb Cell Fact 2016; 15:119. [PMID: 27370777 PMCID: PMC4930576 DOI: 10.1186/s12934-016-0517-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/22/2016] [Indexed: 11/10/2022] Open
Abstract
Hyaluronic acid, or HA, is a rigid and linear biopolymer belonging to the class of the glycosaminoglycans, and composed of repeating units of the monosaccharides glucuronic acid and N-acetylglucosamine. HA has multiple important functions in the human body, due to its properties such as bio-compatibility, lubricity and hydrophilicity, it is widely applied in the biomedical, food, health and cosmetic fields. The growing interest in this molecule has motivated the discovery of new ways of obtaining it. Traditionally, HA has been extracted from rooster comb-like animal tissues. However, due to legislation laws HA is now being produced by bacterial fermentation using Streptococcus zooepidemicus, a natural producer of HA, despite it being a pathogenic microorganism. With the expansion of new genetic engineering technologies, the use of organisms that are non-natural producers of HA has also made it possible to obtain such a polymer. Most of the published reviews have focused on HA formulation and its effects on different body tissues, whereas very few of them describe the microbial basis of HA production. Therefore, for the first time this review has compiled the molecular and genetic bases for natural HA production in microorganisms together with the main strategies employed for heterologous production of HA.
Collapse
Affiliation(s)
- Juliana Davies de Oliveira
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, CEP 70.790-160, Brazil
| | - Lucas Silva Carvalho
- Integra Bioprocessos e Análises, Campus Universitário Darcy Ribeiro, Edifício CDT, Sala AT-36/37, Brasília, DF, CEP 70.904-970, Brazil
| | - Antônio Milton Vieira Gomes
- Grupo de Engenharia Metabólica Aplicada a Bioprocessos, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF, CEP 70.790-900, Brazil
| | - Lúcio Rezende Queiroz
- Grupo de Engenharia Metabólica Aplicada a Bioprocessos, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF, CEP 70.790-900, Brazil
| | - Beatriz Simas Magalhães
- Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, CEP 70.790-160, Brazil.,Integra Bioprocessos e Análises, Campus Universitário Darcy Ribeiro, Edifício CDT, Sala AT-36/37, Brasília, DF, CEP 70.904-970, Brazil
| | - Nádia Skorupa Parachin
- Grupo de Engenharia Metabólica Aplicada a Bioprocessos, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF, CEP 70.790-900, Brazil.
| |
Collapse
|
33
|
Sze JH, Brownlie JC, Love CA. Biotechnological production of hyaluronic acid: a mini review. 3 Biotech 2016; 6:67. [PMID: 28330137 PMCID: PMC4754297 DOI: 10.1007/s13205-016-0379-9] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Hyaluronic acid (HA) is a polysaccharide found in the extracellular matrix of vertebrate epithelial, neural and connective tissues. Due to the high moisture retention, biocompatibility and viscoelasticity properties of this polymer, HA has become an important component of major pharmaceutical, biomedical and cosmetic products with high commercial value worldwide. Currently, large scale production of HA involves extraction from animal tissues as well as the use of bacterial expression systems in Streptococci. However, due to concerns over safety, alternative sources of HA have been pursued which include the use of endotoxin-free microorganisms such as Bacilli and Escherichia coli. In this review, we explore current knowledge of biosynthetic enzymes that produce HA, how these systems have been used commercially to produce HA and how the challenges of producing HA cheaply and safely are being addressed.
Collapse
Affiliation(s)
- Jun Hui Sze
- School of Natural Sciences, Griffith University, Nathan, QLD, 4111, Australia
| | - Jeremy C Brownlie
- School of Natural Sciences, Griffith University, Nathan, QLD, 4111, Australia
- Environmental Futures Research Institute, Griffith University, Nathan, QLD, 4111, Australia
| | - Christopher A Love
- School of Natural Sciences, Griffith University, Nathan, QLD, 4111, Australia.
- Eskitis Institute for Drug Discovery, Griffith University, Nathan, QLD, 4111, Australia.
| |
Collapse
|
34
|
Malitsky S, Ziv C, Rosenwasser S, Zheng S, Schatz D, Porat Z, Ben-Dor S, Aharoni A, Vardi A. Viral infection of the marine alga Emiliania huxleyi triggers lipidome remodeling and induces the production of highly saturated triacylglycerol. THE NEW PHYTOLOGIST 2016; 210:88-96. [PMID: 26856244 DOI: 10.1111/nph.13852] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 12/15/2015] [Indexed: 05/04/2023]
Abstract
Viruses that infect marine photosynthetic microorganisms are major ecological and evolutionary drivers of microbial food webs, estimated to turn over more than a quarter of the total photosynthetically fixed carbon. Viral infection of the bloom-forming microalga Emiliania huxleyi induces the rapid remodeling of host primary metabolism, targeted towards fatty acid metabolism. We applied a liquid chromatography-mass spectrometry (LC-MS)-based lipidomics approach combined with imaging flow cytometry and gene expression profiling to explore the impact of viral-induced metabolic reprogramming on lipid composition. Lytic viral infection led to remodeling of the cellular lipidome, by predominantly inducing the biosynthesis of highly saturated triacylglycerols (TAGs), coupled with a significant accumulation of neutral lipids within lipid droplets. Furthermore, TAGs were found to be a major component (77%) of the lipidome of isolated virions. Interestingly, viral-induced TAGs were significantly more saturated than TAGs produced under nitrogen starvation. This study highlights TAGs as major products of the viral-induced metabolic reprogramming during the host-virus interaction and indicates a selective mode of membrane recruitment during viral assembly, possibly by budding of the virus from specialized subcellular compartments. These findings provide novel insights into the role of viruses infecting microalgae in regulating metabolism and energy transfer in the marine environment and suggest their possible biotechnological application in biofuel production.
Collapse
Affiliation(s)
- Sergey Malitsky
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Carmit Ziv
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shilo Rosenwasser
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shuning Zheng
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Daniella Schatz
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ziv Porat
- Biological Services Department, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shifra Ben-Dor
- Biological Services Department, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Asaph Aharoni
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Assaf Vardi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
35
|
Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev 2016; 97:4-27. [PMID: 26562801 DOI: 10.1016/j.addr.2015.11.001] [Citation(s) in RCA: 1547] [Impact Index Per Article: 171.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network composed of collagens, proteoglycans/glycosaminoglycans, elastin, fibronectin, laminins, and several other glycoproteins. Matrix components bind each other as well as cell adhesion receptors forming a complex network into which cells reside in all tissues and organs. Cell surface receptors transduce signals into cells from ECM, which regulate diverse cellular functions, such as survival, growth, migration, and differentiation, and are vital for maintaining normal homeostasis. ECM is a highly dynamic structural network that continuously undergoes remodeling mediated by several matrix-degrading enzymes during normal and pathological conditions. Deregulation of ECM composition and structure is associated with the development and progression of several pathologic conditions. This article emphasizes in the complex ECM structure as to provide a better understanding of its dynamic structural and functional multipotency. Where relevant, the implication of the various families of ECM macromolecules in health and disease is also presented.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Chrysostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece; Division of Medical Protein Chemistry, Department of Translational Medicine Malmö, Lund University, S-20502 Malmö, Sweden
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
36
|
The Autonomous Glycosylation of Large DNA Viruses. Int J Mol Sci 2015; 16:29315-28. [PMID: 26690138 PMCID: PMC4691112 DOI: 10.3390/ijms161226169] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 11/17/2022] Open
Abstract
Glycosylation of surface molecules is a key feature of several eukaryotic viruses, which use the host endoplasmic reticulum/Golgi apparatus to add carbohydrates to their nascent glycoproteins. In recent years, a newly discovered group of eukaryotic viruses, belonging to the Nucleo-Cytoplasmic Large DNA Virus (NCLDV) group, was shown to have several features that are typical of cellular organisms, including the presence of components of the glycosylation machinery. Starting from initial observations with the chlorovirus PBCV-1, enzymes for glycan biosynthesis have been later identified in other viruses; in particular in members of the Mimiviridae family. They include both the glycosyltransferases and other carbohydrate-modifying enzymes and the pathways for the biosynthesis of the rare monosaccharides that are found in the viral glycan structures. These findings, together with genome analysis of the newly-identified giant DNA viruses, indicate that the presence of glycogenes is widespread in several NCLDV families. The identification of autonomous viral glycosylation machinery leads to many questions about the origin of these pathways, the mechanisms of glycan production, and eventually their function in the viral replication cycle. The scope of this review is to highlight some of the recent results that have been obtained on the glycosylation systems of the large DNA viruses, with a special focus on the enzymes involved in nucleotide-sugar production.
Collapse
|
37
|
Bi Y, Hubbard C, Purushotham P, Zimmer J. Insights into the structure and function of membrane-integrated processive glycosyltransferases. Curr Opin Struct Biol 2015; 34:78-86. [PMID: 26342143 PMCID: PMC4684724 DOI: 10.1016/j.sbi.2015.07.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 12/23/2022]
Abstract
Complex carbohydrates perform essential functions in life, including energy storage, cell signaling, protein targeting, quality control, as well as supporting cell structure and stability. Extracellular polysaccharides (EPS) represent mainly structural polymers and are found in essentially all kingdoms of life. For example, EPS are important biofilm and capsule components in bacteria, represent major constituents in cell walls of fungi, algae, arthropods and plants, and modulate the extracellular matrix in vertebrates. Different mechanisms evolved by which EPS are synthesized. Here, we review the structures and functions of membrane-integrated processive glycosyltransferases (GTs) implicated in the synthesis and secretion of chitin, alginate, hyaluronan and poly-N-acetylglucosamine (PNAG).
Collapse
Affiliation(s)
- Yunchen Bi
- Center for Membrane Biology, Department of Molecular Physiology and Biological Physics, University of Virginia, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, United States
| | - Caitlin Hubbard
- Center for Membrane Biology, Department of Molecular Physiology and Biological Physics, University of Virginia, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, United States
| | - Pallinti Purushotham
- Center for Membrane Biology, Department of Molecular Physiology and Biological Physics, University of Virginia, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, United States
| | - Jochen Zimmer
- Center for Membrane Biology, Department of Molecular Physiology and Biological Physics, University of Virginia, 480 Ray C. Hunt Dr., Charlottesville, VA 22908, United States.
| |
Collapse
|
38
|
Hyaluronan, a truly “youthful” polysaccharide. Its medical applications. ACTA ACUST UNITED AC 2015; 63:32-4. [DOI: 10.1016/j.patbio.2014.05.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 05/12/2014] [Indexed: 11/21/2022]
|
39
|
Marcellin E, Steen JA, Nielsen LK. Insight into hyaluronic acid molecular weight control. Appl Microbiol Biotechnol 2014; 98:6947-56. [DOI: 10.1007/s00253-014-5853-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/22/2014] [Accepted: 05/24/2014] [Indexed: 01/03/2023]
|
40
|
Rosenwasser S, Mausz MA, Schatz D, Sheyn U, Malitsky S, Aharoni A, Weinstock E, Tzfadia O, Ben-Dor S, Feldmesser E, Pohnert G, Vardi A. Rewiring Host Lipid Metabolism by Large Viruses Determines the Fate of Emiliania huxleyi, a Bloom-Forming Alga in the Ocean. THE PLANT CELL 2014; 26:2689-2707. [PMID: 24920329 PMCID: PMC4114960 DOI: 10.1105/tpc.114.125641] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/07/2014] [Accepted: 05/26/2014] [Indexed: 05/21/2023]
Abstract
Marine viruses are major ecological and evolutionary drivers of microbial food webs regulating the fate of carbon in the ocean. We combined transcriptomic and metabolomic analyses to explore the cellular pathways mediating the interaction between the bloom-forming coccolithophore Emiliania huxleyi and its specific coccolithoviruses (E. huxleyi virus [EhV]). We show that EhV induces profound transcriptome remodeling targeted toward fatty acid synthesis to support viral assembly. A metabolic shift toward production of viral-derived sphingolipids was detected during infection and coincided with downregulation of host de novo sphingolipid genes and induction of the viral-encoded homologous pathway. The depletion of host-specific sterols during lytic infection and their detection in purified virions revealed their novel role in viral life cycle. We identify an essential function of the mevalonate-isoprenoid branch of sterol biosynthesis during infection and propose its downregulation as an antiviral mechanism. We demonstrate how viral replication depends on the hijacking of host lipid metabolism during the chemical "arms race" in the ocean.
Collapse
Affiliation(s)
- Shilo Rosenwasser
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michaela A Mausz
- Institute of Inorganic and Analytical Chemistry/Bioorganic Analytics, Friedrich Schiller University Jena, 07743 Jena, Germany Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany
| | - Daniella Schatz
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Uri Sheyn
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sergey Malitsky
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Asaph Aharoni
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal Weinstock
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Oren Tzfadia
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ester Feldmesser
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Georg Pohnert
- Institute of Inorganic and Analytical Chemistry/Bioorganic Analytics, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Assaf Vardi
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
41
|
Chen XW, Kang LH, Ding D, Liu Q, Wang JX, Kang CJ. Characterization of a 2-Cys peroxiredoxin IV in Marsupenaeus japonicus (kuruma shrimp) and its role in the anti-viral immunity. FISH & SHELLFISH IMMUNOLOGY 2013; 35:1848-1857. [PMID: 24056278 DOI: 10.1016/j.fsi.2013.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 09/07/2013] [Accepted: 09/09/2013] [Indexed: 06/02/2023]
Abstract
Accumulating evidence suggests that peroxiredoxins (Prx) are key molecules in the pathogenesis of various infectious diseases and are potential therapeutic targets for major diseases such as cancers. In this study, we report a peroxiredoxin IV (Prx IV) in Marsupenaeus japonicus, designated as MjPrx IV, which exhibited peroxidase activity and participated in the anti-white spot syndrome virus (WSSV) immune response. MjPrx IV is a 245-amino acid polypeptide with a predicted 19-amino acid signal peptide, an Ahpc-TSA domain, and a 1-Cys PrxC domain. Phylogenetic analysis revealed that the protein belongs to the Prx IV subfamily. MjPrx IV transcripts were detected in the gills, hepatopancreas, heart, stomach, ovaries, spermary, and intestine tissues, and are upregulated in the gonads, gills and hemocytes of shrimp after WSSV challenge. The mature MjPrx IV peptide was recombinantly expressed in an Escherichia coli system. The protein exhibited peroxidase activity. Furthermore, dsRNA suppression of MjPrx IV increased WSSV replication in shrimp, whereas rMjPrx IV injection into shrimp decreased WSSV replication. These data suggest that MjPrx IV has an important role in shrimp antiviral immunity. To our knowledge, this study is the first to report a shrimp Prx IV that has anti-WSSV activity.
Collapse
Affiliation(s)
- Xiao-Wei Chen
- The Key Laboratory of Plant Cell Engineering and Germplasm Innovation of Ministry of Education, School of Life Sciences, Shandong University, 27 Shanda South Road, Jinan, Shandong 250100, China; Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 27 Shanda South Road, Jinan, Shandong 250100, China
| | | | | | | | | | | |
Collapse
|
42
|
Tlustá M, Krahulec J, Pepeliaev S, Franke L, Cerný Z, Jílková J. Production of hyaluronic acid by mutant strains of group C Streptococcus. Mol Biotechnol 2013. [PMID: 23180221 DOI: 10.1007/s12033-012-9622-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This study addresses the influence of upstream region sequence on the strength of has operon promoter in highly encapsulated S. equi subsp. zooepidemicus (SEZ). For this purpose, seven different strains were constructed. Each strain carries a point mutation in one of the following positions upstream of the has promoter: -43, -44, -49, and -50 bp. To facilitate measuring of the recombinant promoter relative strength, ß-glucuronidase gene was used as a reporter gene. Three mutations located in positions -49 and -50: AT, GT, and AG, positively impacted has promoter strength when compared to the wild type sequence GG. Conversely, two other mutations: TG and TT, exhibited a slight inhibitory effect. Further, three different strains carrying chromosomal mutations in the has promoter region were constructed. In two cases, the has operon is under the control of a stronger promoter and in the third strain the has operon is controlled by a weaker promoter. The laboratory fermenter scale cultivations confirmed the increase of hyaluronan yields for SEZPhasAG and SEZPhas2G, resulting 116 and 105 %, respectively. As expected, the yield of the hyaluronic acid of SEZPhas2B strain fell to 41 %.
Collapse
Affiliation(s)
- Marcela Tlustá
- Contipro Biotech s r.o., Dolní Dobrouč 401, Czech Republic.
| | | | | | | | | | | |
Collapse
|
43
|
BcsA and BcsB form the catalytically active core of bacterial cellulose synthase sufficient for in vitro cellulose synthesis. Proc Natl Acad Sci U S A 2013; 110:17856-61. [PMID: 24127606 DOI: 10.1073/pnas.1314063110] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cellulose is a linear extracellular polysaccharide. It is synthesized by membrane-embedded glycosyltransferases that processively polymerize UDP-activated glucose. Polymer synthesis is coupled to membrane translocation through a channel formed by the cellulose synthase. Although eukaryotic cellulose synthases function in macromolecular complexes containing several different enzyme isoforms, prokaryotic synthases associate with additional subunits to bridge the periplasm and the outer membrane. In bacteria, cellulose synthesis and translocation is catalyzed by the inner membrane-associated bacterial cellulose synthase (Bcs)A and BcsB subunits. Similar to alginate and poly-β-1,6 N-acetylglucosamine, bacterial cellulose is implicated in the formation of sessile bacterial communities, termed biofilms, and its synthesis is likewise stimulated by cyclic-di-GMP. Biochemical studies of exopolysaccharide synthesis are hampered by difficulties in purifying and reconstituting functional enzymes. We demonstrate robust in vitro cellulose synthesis reconstituted from purified BcsA and BcsB proteins from Rhodobacter sphaeroides. Although BcsA is the catalytically active subunit, the membrane-anchored BcsB subunit is essential for catalysis. The purified BcsA-B complex produces cellulose chains of a degree of polymerization in the range 200-300. Catalytic activity critically depends on the presence of the allosteric activator cyclic-di-GMP, but is independent of lipid-linked reactants. Our data reveal feedback inhibition of cellulose synthase by UDP but not by the accumulating cellulose polymer and highlight the strict substrate specificity of cellulose synthase for UDP-glucose. A truncation analysis of BcsB localizes the region required for activity of BcsA within its C-terminal membrane-associated domain. The reconstituted reaction provides a foundation for the synthesis of biofilm exopolysaccharides, as well as its activation by cyclic-di-GMP.
Collapse
|
44
|
Rakkhumkaew N, Shibatani S, Kawasaki T, Fujie M, Yamada T. Hyaluronan synthesis in cultured tobacco cells (BY-2) expressing a chlorovirus enzyme: cytological studies. Biotechnol Bioeng 2013; 110:1174-9. [PMID: 23404209 DOI: 10.1002/bit.24783] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 10/15/2012] [Accepted: 10/30/2012] [Indexed: 11/06/2022]
Abstract
Extraction of hyaluronan from animals or microbial fermentation has risks including contamination with pathogens and microbial toxins. In this work, tobacco cultured-cells (BY-2) were successfully transformed with a chloroviral hyaluronan synthase (cvHAS) gene to produce hyaluronan. Cytological studies revealed accumulation of HA on the cells, and also in subcellular fractions (protoplasts, miniplasts, vacuoplasts, and vacuoles). Transgenic BY-2 cells harboring a vSPO-cvHAS construct containing the vacuolar targeting signal of sporamin connected to the N-terminus of cvHAS accumulated significant amounts of HA in vacuoles. These results suggested that cvHAS successfully functions on the vacuolar membrane and synthesizes/transports HA into vacuoles. Efficient synthesis of HA using this system provides a new method for practical production of HA.
Collapse
Affiliation(s)
- Numfon Rakkhumkaew
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8530, Japan
| | | | | | | | | |
Collapse
|
45
|
Abstract
Hyaluronan is a polysaccharide with multiple functions in the human body being involved in creating flexible and protective layers in tissues and in many signalling pathways during embryonic development, wound healing, inflammation, and cancer. Hyaluronan is an important component of active pharmaceutical ingredients for treatment of, for example, arthritis and osteoarthritis, and its commercial value far exceeds that of other microbial extracellular polysaccharides. Traditionally hyaluronan is extracted from animal waste which is a well-established process now. However, biotechnological synthesis of biopolymers provides a wealth of new possibilities. Therefore, genetic/metabolic engineering has been applied in the area of tailor-made hyaluronan synthesis. Another approach is the controlled artificial (in vitro) synthesis of hyaluronan by enzymes. Advantage of using microbial and enzymatic synthesis for hyaluronan production is the simpler downstream processing and a reduced risk of viral contamination. In this paper an overview of the different methods used to produce hyaluronan is presented. Emphasis is on the advancements made in the field of the synthesis of bioengineered hyaluronan.
Collapse
|
46
|
Rakkhumkaew N, Kawasaki T, Fujie M, Yamada T. Prolonged synthesis of hyaluronan by Chlorella cells infected with chloroviruses. J Biosci Bioeng 2013; 115:527-31. [PMID: 23273909 DOI: 10.1016/j.jbiosc.2012.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/11/2012] [Accepted: 11/15/2012] [Indexed: 10/27/2022]
Abstract
Hyaluronan (HA) synthesis by microalgal Chlorella cells in combination with chloroviruses represents a unique eco-friendly process for converting solar energy and CO2 into useful materials. However, at the final stage of viral infection, infected host cells are completely lysed, and thus HA should be harvested before cell lysis. In the current study, two methods were investigated to improve the yield of HA: (i) adopting slow-growing chlorovirus isolates and (ii) modification of the virus replication process using an inhibitor of DNA synthesis, aphidicolin. Compared with Paramecium bursaria Chlorella virus type 1 (PBCV-1), the prototype chlorovirus, slow-growing virus isolates (CVO1 and CVTS1) produced a 1.5 times higher concentration of HA in infected Chlorella cultures. Furthermore, addition of aphidicolin, an inhibitor of DNA synthesis, delayed virus replication and increased the final concentration of HA 1.5-fold that of cultures without the addition of aphidicolin. Therefore, a 2- to 3-fold increase in the yield of HA by the Chlorella-virus system was attained by using slow-growing viral isolates and the addition of aphidicolin.
Collapse
Affiliation(s)
- Numfon Rakkhumkaew
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8530, Japan
| | | | | | | |
Collapse
|
47
|
Williamson SJ, Allen LZ, Lorenzi HA, Fadrosh DW, Brami D, Thiagarajan M, McCrow JP, Tovchigrechko A, Yooseph S, Venter JC. Metagenomic exploration of viruses throughout the Indian Ocean. PLoS One 2012; 7:e42047. [PMID: 23082107 PMCID: PMC3474794 DOI: 10.1371/journal.pone.0042047] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 07/02/2012] [Indexed: 11/20/2022] Open
Abstract
The characterization of global marine microbial taxonomic and functional diversity is a primary goal of the Global Ocean Sampling Expedition. As part of this study, 19 water samples were collected aboard the Sorcerer II sailing vessel from the southern Indian Ocean in an effort to more thoroughly understand the lifestyle strategies of the microbial inhabitants of this ultra-oligotrophic region. No investigations of whole virioplankton assemblages have been conducted on waters collected from the Indian Ocean or across multiple size fractions thus far. Therefore, the goals of this study were to examine the effect of size fractionation on viral consortia structure and function and understand the diversity and functional potential of the Indian Ocean virome. Five samples were selected for comprehensive metagenomic exploration; and sequencing was performed on the microbes captured on 3.0-, 0.8- and 0.1 µm membrane filters as well as the viral fraction (<0.1 µm). Phylogenetic approaches were also used to identify predicted proteins of viral origin in the larger fractions of data from all Indian Ocean samples, which were included in subsequent metagenomic analyses. Taxonomic profiling of viral sequences suggested that size fractionation of marine microbial communities enriches for specific groups of viruses within the different size classes and functional characterization further substantiated this observation. Functional analyses also revealed a relative enrichment for metabolic proteins of viral origin that potentially reflect the physiological condition of host cells in the Indian Ocean including those involved in nitrogen metabolism and oxidative phosphorylation. A novel classification method, MGTAXA, was used to assess virus-host relationships in the Indian Ocean by predicting the taxonomy of putative host genera, with Prochlorococcus, Acanthochlois and members of the SAR86 cluster comprising the most abundant predictions. This is the first study to holistically explore virioplankton dynamics across multiple size classes and provides unprecedented insight into virus diversity, metabolic potential and virus-host interactions.
Collapse
Affiliation(s)
- Shannon J Williamson
- Microbial and Environmental Genomics, J. Craig Venter Institute, San Diego, California, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hubbard C, McNamara JT, Azumaya C, Patel MS, Zimmer J. The Hyaluronan Synthase Catalyzes the Synthesis and Membrane Translocation of Hyaluronan. J Mol Biol 2012; 418:21-31. [DOI: 10.1016/j.jmb.2012.01.053] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/25/2012] [Accepted: 01/31/2012] [Indexed: 12/25/2022]
|
49
|
DeAngelis PL. Glycosaminoglycan polysaccharide biosynthesis and production: today and tomorrow. Appl Microbiol Biotechnol 2012; 94:295-305. [PMID: 22391966 DOI: 10.1007/s00253-011-3801-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 11/29/2011] [Accepted: 12/01/2011] [Indexed: 10/28/2022]
Abstract
Glycosaminoglycans [GAGs] are essential heteropolysaccharides in vertebrate tissues that are also, in certain cases, employed as virulence factors by microbes. Hyaluronan [HA], heparin, and chondroitin sulfate [CS] are GAGs currently used in various medical applications and together are multi-billion dollar products thus targets for production by animal-free manufacture. By using bacteria as the source of GAGs, the pathogen's sword may be converted into a plowshare to help avoid potential liabilities springing from the use of animal-derived GAGs including adventitious agents (e.g., prions, pathogens), antigenicity, degradation of the environment, and depletion of endangered species. HA from microbes, which have a chemical structure identical to human HA, has already been commercialized and sold at the ton-scale. Substantial progress towards microbial heparin and CS has been made, but these vertebrate polymers are more complicated structurally than the unsulfated bacterial polysaccharide precursors thus require additional processing steps. This review provides an overview of GAG structure, medical applications, microbial biosynthesis, and the state of bacterial GAG production systems. Representatives of all glycosyltransferase enzymes that polymerize the sugar chains of the three main GAGs have been identified and serve as the core technology to harness, but the proteins involved in sugar precursor formation and chain export steps of biosynthesis are also essential to the GAG production process. In addition, this review discusses future directions and potential important issues. Overall, this area is poised to make great headway to produce safer (both increased purity and more secure supply chains) non-animal GAG-based therapeutics.
Collapse
Affiliation(s)
- Paul L DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA.
| |
Collapse
|
50
|
Van Etten JL, Dunigan DD. Chloroviruses: not your everyday plant virus. TRENDS IN PLANT SCIENCE 2012; 17:1-8. [PMID: 22100667 PMCID: PMC3259250 DOI: 10.1016/j.tplants.2011.10.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/20/2011] [Accepted: 10/25/2011] [Indexed: 05/29/2023]
Abstract
Viruses infecting higher plants are among the smallest viruses known and typically have four to ten protein-encoding genes. By contrast, many viruses that infect algae (classified in the virus family Phycodnaviridae) are among the largest viruses found to date and have up to 600 protein-encoding genes. This brief review focuses on one group of plaque-forming phycodnaviruses that infect unicellular chlorella-like green algae. The prototype chlorovirus PBCV-1 has more than 400 protein-encoding genes and 11 tRNA genes. About 40% of the PBCV-1 encoded proteins resemble proteins of known function including many that are completely unexpected for a virus. In many respects, chlorovirus infection resembles bacterial infection by tailed bacteriophages.
Collapse
Affiliation(s)
- James L Van Etten
- Department of Plant Pathology and Nebraska Center for Virology, University of Nebraska, Lincoln, NE 68583-0900, USA.
| | | |
Collapse
|