Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 6961, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37907464

ABSTRACT

The spirochete bacterial pathogen Borrelia (Borreliella) burgdorferi (Bbu) affects more than 10% of the world population and causes Lyme disease in about half a million people in the US annually. Therapy for Lyme disease includes antibiotics that target the Bbu ribosome. Here we present the structure of the Bbu 70S ribosome obtained by single particle cryo-electron microscopy at 2.9 Å resolution, revealing a bound hibernation promotion factor protein and two genetically non-annotated ribosomal proteins bS22 and bL38. The ribosomal protein uL30 in Bbu has an N-terminal α-helical extension, partly resembling the mycobacterial bL37 protein, suggesting evolution of bL37 and a shorter uL30 from a longer uL30 protein. Its analogy to proteins uL30m and mL63 in mammalian mitochondrial ribosomes also suggests a plausible evolutionary pathway for expansion of protein content in mammalian mitochondrial ribosomes. Computational binding free energy predictions for antibiotics reflect subtle distinctions in antibiotic-binding sites in the Bbu ribosome. Discovery of these features in the Bbu ribosome may enable better ribosome-targeted antibiotic design for Lyme disease treatment.


Subject(s)
Bacterial Proteins , Lyme Disease , Animals , Humans , Cryoelectron Microscopy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ribosomes/metabolism , Ribosomal Proteins/metabolism , Anti-Bacterial Agents/metabolism , Mammals/metabolism
3.
Proc Natl Acad Sci U S A ; 120(27): e2301549120, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37364114

ABSTRACT

Modern infectious disease outbreaks often involve changes in host tropism, the preferential adaptation of pathogens to specific hosts. The Lyme disease-causing bacterium Borrelia burgdorferi (Bb) is an ideal model to investigate the molecular mechanisms of host tropism, because different variants of these tick-transmitted bacteria are distinctly maintained in rodents or bird reservoir hosts. To survive in hosts and escape complement-mediated immune clearance, Bb produces the outer surface protein CspZ that binds the complement inhibitor factor H (FH) to facilitate bacterial dissemination in vertebrates. Despite high sequence conservation, CspZ variants differ in human FH-binding ability. Together with the FH polymorphisms between vertebrate hosts, these findings suggest that minor sequence variation in this bacterial outer surface protein may confer dramatic differences in host-specific, FH-binding-mediated infectivity. We tested this hypothesis by determining the crystal structure of the CspZ-human FH complex, and identifying minor variation localized in the FH-binding interface yielding bird and rodent FH-specific binding activity that impacts infectivity. Swapping the divergent region in the FH-binding interface between rodent- and bird-associated CspZ variants alters the ability to promote rodent- and bird-specific early-onset dissemination. We further linked these loops and respective host-specific, complement-dependent phenotypes with distinct CspZ phylogenetic lineages, elucidating evolutionary mechanisms driving host tropism emergence. Our multidisciplinary work provides a novel molecular basis for how a single, short protein motif could greatly modulate pathogen host tropism.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Animals , Humans , Immune Evasion/genetics , Phylogeny , Viral Tropism , Lyme Disease/microbiology , Bacterial Proteins/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Complement System Proteins/genetics , Membrane Proteins/metabolism
4.
bioRxiv ; 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37131667

ABSTRACT

The spirochete bacterial pathogen Borrelia ( Borreliella) burgdorferi ( Bbu ) affects more than 10% of the world population and causes Lyme disease in about half a million people in the US annually. Therapy for Lyme disease includes antibiotics that target the Bbu ribosome. We determined the structure of the Bbu 70S ribosome by single particle cryo-electron microscopy (cryo-EM) at a resolution of 2.9 Å, revealing its distinctive features. In contrast to a previous study suggesting that the single hibernation promoting factor protein present in Bbu (bbHPF) may not bind to its ribosome, our structure reveals a clear density for bbHPF bound to the decoding center of the small ribosomal 30S subunit. The 30S subunit has a non-annotated ribosomal protein, bS22, that has been found only in mycobacteria and Bacteroidetes so far. The protein bL38, recently discovered in Bacteroidetes, is also present in the Bbu large 50S ribosomal subunit. The protein bL37, previously seen only in mycobacterial ribosomes, is replaced by an N-terminal α-helical extension of uL30, suggesting that the two bacterial ribosomal proteins uL30 and bL37 may have evolved from one longer uL30 protein. The longer uL30 protein interacts with both the 23S rRNA and the 5S rRNA, is near the peptidyl transferase center (PTC), and could impart greater stability to this region. Its analogy to proteins uL30m and mL63 in mammalian mitochondrial ribosomes also suggests a plausible evolutionary pathway for expansion of protein content in mammalian mitochondrial ribosomes. Computational binding free energies are predicted for antibiotics, bound to the decoding center or PTC and are in clinical use for Lyme disease, that account for subtle distinctions in antibiotic-binding regions in the Bbu ribosome structure. Besides revealing unanticipated structural and compositional features for the Bbu ribosome, our study thus provides groundwork to enable ribosome-targeted antibiotic design for more effective treatment of Lyme disease.

5.
Ticks Tick Borne Dis ; 14(1): 102081, 2023 01.
Article in English | MEDLINE | ID: mdl-36403322

ABSTRACT

In North America, Lyme disease is primarily caused by the spirochetal bacterium Borrelia burgdorferi sensu stricto (Bb), which is transmitted between multiple vertebrate hosts and ixodid ticks, and is a model commonly used to study host-pathogen interactions. While Bb is consistently observed in its mammalian and avian reservoirs, the bacterium is rarely isolated from North American reptiles. Two closely related lizard species, the eastern fence lizard (Sceloporus undulatus) and the western fence lizard (Sceloporus occidentalis), are examples of reptiles parasitized by Ixodes ticks. Vertebrates are known to generate complement as an innate defense mechanism, which can be activated before Bb disseminate to distal tissues. Complement from western fence lizards has proven lethal against one Bb strain, implying the role of complement in making those lizards unable to serve as hosts to Bb. However, Bb DNA is occasionally identified in distal tissues of field-collected eastern fence lizards, suggesting some Bb strains may overcome complement-mediated clearance in these lizards. These findings raise questions regarding the role of complement and its impact on Bb interactions with North American lizards. In this study, we found Bb seropositivity in a small population of wild-caught eastern fence lizards and observed Bb strain-specific survivability in lizard sera. We also found that a Bb outer surface protein, OspE, from Bb strains viable in sera, promotes lizard serum survivability and binds to a complement inhibitor, factor H, from eastern fence lizards. Our data thus identify bacterial and host determinants of eastern fence lizard complement evasion, providing insights into the role of complement influencing Bb interactions with North American lizards.


Subject(s)
Antigens, Bacterial , Bacterial Outer Membrane Proteins , Borrelia burgdorferi , Complement System Proteins , Immune Evasion , Lipoproteins , Lizards , Lyme Disease , Animals , Borrelia burgdorferi/immunology , Lizards/blood , Lizards/immunology , Lizards/microbiology , North America , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/blood , Bacterial Outer Membrane Proteins/immunology , Lipoproteins/blood , Lipoproteins/immunology , Complement System Proteins/immunology , Lyme Disease/blood , Lyme Disease/immunology , Lyme Disease/microbiology , Lyme Disease/virology
6.
mSystems ; 7(4): e0048822, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35938719

ABSTRACT

Host association-the selective adaptation of pathogens to specific host species-evolves through constant interactions between host and pathogens, leaving a lot yet to be discovered on immunological mechanisms and genomic determinants. The causative agents of Lyme disease (LD) are spirochete bacteria composed of multiple species of the Borrelia burgdorferi sensu lato complex, including B. burgdorferi (Bb), the main LD pathogen in North America-a useful model for the study of mechanisms underlying host-pathogen association. Host adaptation requires pathogens' ability to evade host immune responses, such as complement, the first-line innate immune defense mechanism. We tested the hypothesis that different host-adapted phenotypes among Bb strains are linked to polymorphic loci that confer complement evasion traits in a host-specific manner. We first examined the survivability of 20 Bb strains in sera in vitro and/or bloodstream and tissues in vivo from rodent and avian LD models. Three groups of complement-dependent host-association phenotypes emerged. We analyzed complement-evasion genes, identified a priori among all strains and sequenced and compared genomes for individual strains representing each phenotype. The evolutionary history of ospC loci is correlated with host-specific complement-evasion phenotypes, while comparative genomics suggests that several gene families and loci are potentially involved in host association. This multidisciplinary work provides novel insights into the functional evolution of host-adapted phenotypes, building a foundation for further investigation of the immunological and genomic determinants of host association. IMPORTANCE Host association is the phenotype that is commonly found in many pathogens that preferential survive in particular hosts. The Lyme disease (LD)-causing agent, B. burgdorferi (Bb), is an ideal model to study host association, as Bb is mainly maintained in nature through rodent and avian hosts. A widespread yet untested concept posits that host association in Bb strains is linked to Bb functional genetic variation conferring evasion to complement, an innate defense mechanism in vertebrate sera. Here, we tested this concept by grouping 20 Bb strains into three complement-dependent host-association phenotypes based on their survivability in sera and/or bloodstream and distal tissues in rodent and avian LD models. Phylogenomic analysis of these strains further correlated several gene families and loci, including ospC, with host-specific complement-evasion phenotypes. Such multifaceted studies thus pave the road to further identify the determinants of host association, providing mechanistic insights into host-pathogen interaction.


Subject(s)
Borrelia burgdorferi , Borrelia , Lyme Disease , Humans , Phylogeny , Lyme Disease/genetics , Borrelia burgdorferi/genetics , Complement System Proteins/genetics
7.
Infect Immun ; 90(7): e0006222, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35861564

ABSTRACT

Transmitted by ticks, the bacterium Borrelia burgdorferi sensu lato is the causative agent of Lyme disease (LD), the most common vector-borne disease in the Northern hemisphere. No effective vaccines are currently available. B. burgdorferi sensu lato produces the CspZ protein that binds to the complement inhibitor, factor H (FH), promoting evasion of the host complement system. We previously showed that while vaccination with CspZ did not protect mice from B. burgdorferi infection, mice can be protected after immunization with CspZ-Y207A/Y211A (CspZ-YA), a CspZ mutant protein without FH-binding activity. To further study the mechanism of this protection, herein we evaluated both poly- and monoclonal antibodies recognizing CspZ FH-binding or non-FH-binding sites. We found that the anti-CspZ antibodies that recognize the FH-binding sites (i.e., block FH-binding activity) eliminate B. burgdorferi sensu lato in vitro more efficiently than those that bind to the non-FH-binding sites, and passive inoculation with anti-FH-binding site antibodies eradicated B. burgdorferi sensu lato in vivo. Antibodies against non-FH-binding sites did not have the same effect. These results emphasize the importance of CspZ FH-binding sites in triggering a protective antibody response against B. burgdorferi sensu lato in future LD vaccines.


Subject(s)
Borrelia burgdorferi Group , Borrelia , Ixodes , Lyme Disease , Animals , Antibodies , Binding Sites , Complement Factor H , Epitopes , Ixodes/microbiology , Lyme Disease/microbiology , Mice
8.
Proc Biol Sci ; 289(1969): 20212087, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35193398

ABSTRACT

Predicting pathogen emergence and spillover risk requires understanding the determinants of a pathogens' host range and the traits involved in host competence. While host competence is often considered a fixed species-specific trait, it may be variable if pathogens diversify across hosts. Balancing selection can lead to maintenance of pathogen polymorphisms (multiple-niche-polymorphism; MNP). The causative agent of Lyme disease, Borrelia burgdorferi (Bb), provides a model to study the evolution of host adaptation, as some Bb strains defined by their outer surface protein C (ospC) genotype, are widespread in white-footed mice and others are associated with non-rodent vertebrates (e.g. birds). To identify the mechanisms underlying potential strain × host adaptation, we infected American robins and white-footed mice, with three Bb strains of different ospC genotypes. Bb burdens varied by strain in a host-dependent fashion, and strain persistence in hosts largely corresponded to Bb survival at early infection stages and with transmission to larvae (i.e. fitness). Early survival phenotypes are associated with cell adhesion, complement evasion and/or inflammatory and antibody-mediated removal of Bb, suggesting directional selective pressure for host adaptation and the potential role of MNP in maintaining OspC diversity. Our findings will guide future investigations to inform eco-evolutionary models of host adaptation for microparasites.


Subject(s)
Borrelia burgdorferi Group , Borrelia burgdorferi , Lyme Disease , Animals , Borrelia burgdorferi/genetics , Borrelia burgdorferi Group/genetics , Host Adaptation , Peromyscus , Phenotype
9.
Infect Immun ; 88(5)2020 04 20.
Article in English | MEDLINE | ID: mdl-32122944

ABSTRACT

The spirochete Borrelia burgdorferisensu lato is the causative agent of Lyme disease (LD). The spirochetes produce the CspZ protein that binds to a complement regulator, factor H (FH). Such binding downregulates activation of host complement to facilitate spirochete evasion of complement killing. However, vaccination with CspZ does not protect against LD infection. In this study, we demonstrated that immunization with CspZ-YA, a CspZ mutant protein with no FH-binding activity, protected mice from infection by several spirochete genotypes introduced via tick feeding. We found that the sera from CspZ-YA-vaccinated mice more efficiently eliminated spirochetes and blocked CspZ FH-binding activity than sera from CspZ-immunized mice. We also found that vaccination with CspZ, but not CspZ-YA, triggered the production of anti-FH antibodies, justifying CspZ-YA as an LD vaccine candidate. The mechanistic and efficacy information derived from this study provides insights into the development of a CspZ-based LD vaccine.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Complement Factor H/immunology , Lyme Disease/immunology , Ticks/microbiology , Animals , Antibodies/immunology , Binding Sites/immunology , Complement System Proteins/immunology , Female , Humans , Lyme Disease Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
10.
ACS Infect Dis ; 6(3): 503-514, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31961652

ABSTRACT

Lyme disease (LD) is caused by the spirochete Borrelia burgdorferi sensu lato (Bbsl). After transmission to humans by ticks, Bbsl spreads to multiple organs, leading to arthritis, carditis, and neuroborreliosis. No effective prophylaxis against human LD prior to tick exposure is currently available. Thus, a pre-exposure prophylaxis (PrEP) against LD is needed. The establishment of LD bacteria at diverse sites is dictated partly by the binding of Bbsl to proteoglycans (PGs) and glycosaminoglycans (GAGs) in tissues. The drug heparin is structurally similar to these GAGs and inhibits Bbsl attachment to PGs, GAGs, cells, and tissues, suggesting its potential to prevent LD. However, the anticoagulant activity of heparin often results in hemorrhage, hampering the development of this compound as LD PrEP. We have previously synthesized a non-anticoagulant version of heparin (NACH), which was verified for safety in mice and humans. Here, we showed that NACH blocks Bbsl attachment to PGs, GAGs, and mammalian cells. We also found that treating mice with NACH prior to the exposure of ticks carrying Bbsl followed by continuous administration of this compound prevents tissue colonization by Bbsl. Furthermore, NACH-treated mice develop greater levels of IgG and IgM against Bbsl at early stages of infection, suggesting that the upregulation of antibody immune responses may be one of the mechanisms for NACH-mediated LD prevention. This is one of the first studies examining the ability of a heparin-based compound to prevent LD prior to tick exposure. The information presented might also be extended to prevent other infectious diseases agents.


Subject(s)
Borrelia burgdorferi/drug effects , Heparin/administration & dosage , Lyme Disease/prevention & control , Pre-Exposure Prophylaxis , Animals , Female , Heparin/chemistry , Mice , Mice, Inbred BALB C
11.
Vaccine ; 38(4): 942-950, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31727504

ABSTRACT

Outer surface protein A (OspA) is a Borrelia lipoprotein and an established Lyme disease vaccine target. Admixing non-lipidated, recombinant B. burgdorferi OspA with liposomes containing cobalt porphyrin-phospholipid (CoPoP) resulted in rapid, particulate surface display of the conformationally intact antigen. Particleization was serum-stable and led to enhanced antigen uptake in murine macrophages in vitro. Mouse immunization using CoPoP liposomes that also contained a synthetic monophosphoryl lipid A (PHAD) elicited a Th1-biased OspA antibody response with higher IgG production compared to other vaccine adjuvants. Antibodies were reactive with intact B. burgdorferi spirochetes and Borrelia lysates, and induced complement-mediated borreliacidal activity in vitro. One year after initial immunization, mice maintained high levels of circulating borreliacidal antibodies capable of blocking B. burgdorferi transmission from infected ticks to human blood in a feeding chamber.


Subject(s)
Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Lipoproteins/immunology , Lyme Disease Vaccines/administration & dosage , Lyme Disease/prevention & control , Vaccination , Animals , Antibody Formation/immunology , Cobalt/chemistry , Female , Immunogenicity, Vaccine , Liposomes , Lyme Disease/immunology , Lyme Disease Vaccines/immunology , Macrophages/immunology , Mice , Mice, Inbred ICR , Phospholipids/chemistry , Porphyrins/chemistry , Time Factors
12.
Cell Microbiol ; 21(2): e12998, 2019 02.
Article in English | MEDLINE | ID: mdl-30571845

ABSTRACT

Lyme disease, caused by the spirochete Borrelia burgdorferi, is the most common vector-borne disease in the United States and Europe. The spirochetes are transmitted from mammalian and avian reservoir hosts to humans via ticks. Following tick bites, spirochetes colonize the host skin and then disseminate haematogenously to various organs, a process that requires this pathogen to evade host complement, an innate immune defence system. CspZ, a spirochete surface protein, facilitates resistance to complement-mediated killing in vitro by binding to the complement regulator, factor H (FH). Low expression levels of CspZ in spirochetes cultivated in vitro or during initiation of infection in vivo have been a major hurdle in delineating the role of this protein in pathogenesis. Here, we show that treatment of B. burgdorferi with human blood induces CspZ production and enhances resistance to complement. By contrast, a cspZ-deficient mutant and a strain that expressed an FH-nonbinding CspZ variant were impaired in their ability to cause bacteraemia and colonize tissues of mice or quail; virulence of these mutants was however restored in complement C3-deficient mice. These novel findings suggest that FH binding to CspZ facilitates B. burgdorferi complement evasion in vivo and promotes systemic infection in vertebrate hosts.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/immunology , Complement C3/immunology , Lyme Disease/immunology , Membrane Proteins/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/pathogenicity , Complement C3/genetics , Complement Factor H/immunology , Complement Factor H/metabolism , Coturnix , Humans , Ixodes/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
13.
Front Immunol ; 9: 181, 2018.
Article in English | MEDLINE | ID: mdl-29472926

ABSTRACT

The spirochete Borrelia burgdorferi is the causative agent of Lyme disease, the most common tick-borne disease in the US and Europe. No potent human vaccine is currently available. The innate immune complement system is vital to host defense against pathogens, as complement activation on the surface of spirochetes results in bacterial killing. Complement system is inhibited by the complement regulator factor H (FH). To escape killing, B. burgdorferi produces an outer surface protein CspZ that binds FH to inhibit complement activation on the cell surface. Immunization with CspZ alone does not protect mice from infection, which we speculate is because FH-binding cloaks potentially protective epitopes. We modified CspZ by conjugating to virus-like particles (VLP-CspZ) and eliminating FH binding (modified VLP-CspZ) to increase immunogenicity. We observed greater bactericidal antibody titers in mice vaccinated with modified VLP-CspZ: A serum dilution of 1:395 (modified VLP-CspZ) vs 1:143 (VLP-CspZ) yielded 50% borreliacidal activity. Immunizing mice with modified VLP-CspZ cleared spirochete infection, as did passive transfer of elicited antibodies. This work developed a novel Lyme disease vaccine candidate by conjugating CspZ to VLP and eliminating FH-binding ability. Such a strategy of conjugating an antigen to a VLP and eliminating binding to the target ligand can serve as a general model for developing vaccines against other bacterial infectious agents.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement Factor H/immunology , Lyme Disease Vaccines/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Bacterial/administration & dosage , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi , Complement Factor H/genetics , Immunization, Passive , Lyme Disease/immunology , Lyme Disease/prevention & control , Male , Mice , Serum Bactericidal Antibody Assay , Vaccines, Virus-Like Particle/genetics
14.
Front Microbiol ; 8: 328, 2017.
Article in English | MEDLINE | ID: mdl-28303129

ABSTRACT

Lyme disease and relapsing fever are caused by various Borrelia species. Lyme disease borreliae, the most common vector-borne pathogens in both the U.S. and Europe, are transmitted by Ixodes ticks and disseminate from the site of tick bites to tissues leading to erythema migrans skin rash, arthritis, carditis, and neuroborreliosis. Relapsing fever borreliae, carried by ticks and lice, trigger reoccurring fever episodes. Following transmission, spirochetes survive in the blood to induce bacteremia at the early stages of infection, which is thought to promote evasion of the host complement system. The complement system acts as an important innate immune defense mechanism in humans and vertebrates. Upon activation, the cleaved complement components form complexes on the pathogen surface to eventually promote bacteriolysis. The complement system is negatively modulated by a number of functionally diverse regulators to avoid tissue damage. To evade and inhibit the complement system, spirochetes are capable of binding complement components and regulators. Complement inhibition results in bacterial survival in serum (serum resistance) and is thought to promote bloodstream survival, which facilitates spirochete dissemination and disease manifestations. In this review, we discuss current methodologies to elucidate the mechanisms of Borrelia spp. that promote serum resistance and bloodstream survival, as well as novel methods to study factors responsible for bloodstream survival of Lyme disease borreliae that can be applied to relapsing fever borreliae. Understanding the mechanisms these pathogens utilize to evade the complement system will ultimately aid in the development of novel therapeutic strategies and disease prevention to improve human health.

15.
PLoS One ; 11(5): e0155018, 2016.
Article in English | MEDLINE | ID: mdl-27144925

ABSTRACT

Reports from state health departments and the Centers for Disease Control and Prevention indicate that the annual number of reported human vibriosis cases in New England has increased in the past decade. Concurrently, there has been a shift in both the spatial distribution and seasonal detection of Vibrio spp. throughout the region based on limited monitoring data. To determine environmental factors that may underlie these emerging conditions, this study focuses on a long-term database of Vibrio parahaemolyticus concentrations in oyster samples generated from data collected from the Great Bay Estuary, New Hampshire over a period of seven consecutive years. Oyster samples from two distinct sites were analyzed for V. parahaemolyticus abundance, noting significant relationships with various biotic and abiotic factors measured during the same period of study. We developed a predictive modeling tool capable of estimating the likelihood of V. parahaemolyticus presence in coastal New Hampshire oysters. Results show that the inclusion of chlorophyll a concentration to an empirical model otherwise employing only temperature and salinity variables, offers improved predictive capability for modeling the likelihood of V. parahaemolyticus in the Great Bay Estuary.


Subject(s)
Bays/microbiology , Vibrio parahaemolyticus/isolation & purification , Animals , Chlorophyll/metabolism , Chlorophyll A , Environment , Humans , New England , New Hampshire , Ostreidae/microbiology , Salinity , Seasons , Temperature , Vibrio Infections/microbiology , Water Microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...