fbpx
Wikipedia

Biofilm

A biofilm is an syntrophic community of microorganisms in which cells stick to each other and often also to a surface.[2][3][4] These adherent cells become embedded within a slimy extracellular matrix that is composed of extracellular polymeric substances (EPSs).[3][4] The cells within the biofilm produce the EPS components, which are typically a polymeric combination of extracellular polysaccharides, proteins, lipids and DNA.[3][4][5] Because they have three-dimensional structure and represent a community lifestyle for microorganisms, they have been metaphorically described as "cities for microbes".[6][7]

Staphylococcus aureus biofilm on an indwelling catheter
Probable cyanobacteria in the vertical section of a silicified biofilm from the Lower Cretaceous. Very shallow hypersaline environment of the Urgonian carbonate platform of Provence, south eastern France.
IUPAC definition

Aggregate of microorganisms in which cells that are frequently embedded within a self-produced matrix of extracellular polymeric substances (EPSs) adhere to each other and/or to a surface.

  • A biofilm is a system that can be adapted internally to environmental conditions by its inhabitants.
  • The self-produced matrix of extracellular polymeric substances, which is also referred to as slime, is a polymeric conglomeration generally composed of extracellular biopolymers in various structural forms.[1]

Biofilms may form on living (biotic) or non-living (abiotic) surfaces and can be common in natural, industrial, and hospital settings.[4][8] They may constitute a microbiome or be a portion of it. The microbial cells growing in a biofilm are physiologically distinct from planktonic cells of the same organism, which, by contrast, are single cells that may float or swim in a liquid medium.[9] Biofilms can form on the teeth of most animals as dental plaque, where they may cause tooth decay and gum disease.

Microbes form a biofilm in response to a number of different factors,[10] which may include cellular recognition of specific or non-specific attachment sites on a surface, nutritional cues, or in some cases, by exposure of planktonic cells to sub-inhibitory concentrations of antibiotics.[11][12] A cell that switches to the biofilm mode of growth undergoes a phenotypic shift in behavior in which large suites of genes are differentially regulated.[13]

A biofilm may also be considered a hydrogel, which is a complex polymer that contains many times its dry weight in water. Biofilms are not just bacterial slime layers but biological systems; the bacteria organize themselves into a coordinated functional community. Biofilms can attach to a surface such as a tooth or rock, and may include a single species or a diverse group of microorganisms. Subpopulations of cells within the biofilm differentiate to perform various activities for motility, matrix production, and sporulation, supporting the overall success of the biofilm.[14] The biofilm bacteria can share nutrients and are sheltered from harmful factors in the environment, such as desiccation, antibiotics, and a host body's immune system. A biofilm usually begins to form when a free-swimming, planktonic bacterium attaches to a surface.[15][page needed]

Origin and formation edit

Origin of biofilms edit

Biofilms are thought to have arisen during primitive Earth as a defense mechanism for prokaryotes, as the conditions at that time were too harsh for their survival. They can be found very early in Earth's fossil records (about 3.25 billion years ago) as both Archaea and Bacteria, and commonly protect prokaryotic cells by providing them with homeostasis, encouraging the development of complex interactions between the cells in the biofilm.[4]

Formation of biofilms edit

The formation of a biofilm begins with the attachment of free-floating microorganisms to a surface.[9][6] The first colonist bacteria of a biofilm may adhere to the surface initially by the weak van der Waals forces and hydrophobic effects.[16][17] If the colonists are not immediately separated from the surface, they can anchor themselves more permanently using cell adhesion structures such as pili. A unique group of Archaea that inhabit anoxic groundwater have similar structures called hami. Each hamus is a long tube with three hook attachments that are used to attach to each other or to a surface, enabling a community to develop.[18][19] Hyperthermophilic archaeon Pyrobaculum calidifontis produce bundling pili which are homologous to the bacterial TasA filaments, a major component of the extracellular matrix in bacterial biofilms, which contribute to biofilm stability.[20] TasA homologs are encoded by many other archaea, suggesting mechanistic similarities and evolutionary connection between bacterial and archaeal biofilms.[20]

 
Biofilm of golden hydrophobic bacteria; ceiling of Golden Dome Cave, a lava tube in Lava Beds National Monument[21]

Hydrophobicity can also affect the ability of bacteria to form biofilms. Bacteria with increased hydrophobicity have reduced repulsion between the substratum and the bacterium.[22] Some bacteria species are not able to attach to a surface on their own successfully due to their limited motility but are instead able to anchor themselves to the matrix or directly to other, earlier bacteria colonists. Non-motile bacteria cannot recognize surfaces or aggregate together as easily as motile bacteria.[22]

During surface colonization bacteria cells are able to communicate using quorum sensing (QS) products such as N-acyl homoserine lactone (AHL). Once colonization has begun, the biofilm grows by a combination of cell division and recruitment. Polysaccharide matrices typically enclose bacterial biofilms. The matrix exopolysaccharides can trap QS autoinducers within the biofilm to prevent predator detection and ensure bacterial survival.[23] In addition to the polysaccharides, these matrices may also contain material from the surrounding environment, including but not limited to minerals, soil particles, and blood components, such as erythrocytes and fibrin.[22] The final stage of biofilm formation is known as development, and is the stage in which the biofilm is established and may only change in shape and size.[citation needed]

The development of a biofilm may allow for an aggregate cell colony (or colonies) to be increasingly tolerant[24] or resistant to antibiotics. Cell-cell communication or quorum sensing has been shown to be involved in the formation of biofilm in several bacterial species.[25]

Development edit

 
Mature biofilm structure[26]
Biofilm is characterised by heterogenous environment and the presence of a variety of subpopulations. A biofilm structure is composed of metabolically active (both resistant and tolerant) and non-active cells (viable but not culturable cells and persisters) as well as polymer matrix consisting of polysaccharide, extracellular DNA and proteins. Biofilm growth is associated with an escalated level of mutations and horizontal gene transfer which is promoted in due to the packed and dense structure. Bacteria in biofilms communicate by quorum sensing, which activates genes participating in virulence factors production.[26][27]

Biofilms are the product of a microbial developmental process.[28] The process is summarized by five major stages of biofilm development, as shown in the diagram below:[29]

 
Five stages of biofilm development[29]
(1) Initial attachment, (2) Irreversible attachment, (3) Maturation I, (4) Maturation II, and (5) Dispersion. Each stage of development in the diagram is paired with a photomicrograph of a developing P. aeruginosa biofilm. All photomicrographs are shown to the same scale.

Dispersal edit

Dispersal of cells from the biofilm colony is an essential stage of the biofilm life cycle. Dispersal enables biofilms to spread and colonize new surfaces. Enzymes that degrade the biofilm extracellular matrix, such as dispersin B and deoxyribonuclease, may contribute to biofilm dispersal.[30][31] Enzymes that degrade the biofilm matrix may be useful as anti-biofilm agents.[32][33] Evidence has shown that a fatty acid messenger, cis-2-decenoic acid, is capable of inducing dispersion and inhibiting growth of biofilm colonies. Secreted by Pseudomonas aeruginosa, this compound induces cyclo heteromorphic cells in several species of bacteria and the yeast Candida albicans.[34] Nitric oxide has also been shown to trigger the dispersal of biofilms of several bacteria species[35][36] at sub-toxic concentrations. Nitric oxide has potential as a treatment for patients that have chronic infections caused by biofilms.[37]

It was generally assumed that cells dispersed from biofilms immediately go into the planktonic growth phase. However, studies have shown that the physiology of dispersed cells from Pseudomonas aeruginosa biofilms is highly different from that of planktonic and biofilm cells.[38][39] Hence, the dispersal process is a unique stage during the transition from biofilm to planktonic lifestyle in bacteria. Dispersed cells are found to be highly virulent against macrophages and Caenorhabditis elegans, but highly sensitive towards iron stress, as compared with planktonic cells.[38]

Furthermore, Pseudomonas aeruginosa biofilms undergo distinct spatiotemporal dynamics during biofilm dispersal or disassembly, with contrasting consequences in recolonization and disease dissemination.[40] Biofilm dispersal induced bacteria to activate dispersal genes to actively depart from biofilms as single cells at consistent velocities but could not recolonize fresh surfaces. In contrast, biofilm disassembly by degradation of a biofilm exopolysaccharide released immotile aggregates at high initial velocities, enabling the bacteria to recolonize fresh surfaces and cause infections in the hosts efficiently. Hence, biofilm dispersal is more complex than previously thought, where bacterial populations adopting distinct behavior after biofilm departure may be the key to survival of bacterial species and dissemination of diseases.

 
Biofilm dispersal

Properties edit

Biofilms are usually found on solid substrates submerged in or exposed to an aqueous solution, although they can form as floating mats on liquid surfaces and also on the surface of leaves, particularly in high humidity climates. Given sufficient resources for growth, a biofilm will quickly grow to be macroscopic (visible to the naked eye). Biofilms can contain many different types of microorganism, e.g. bacteria, archaea, protozoa, fungi and algae; each group performs specialized metabolic functions. However, some organisms will form single-species films under certain conditions. The social structure (cooperation/competition) within a biofilm depends highly on the different species present.[41]

Extracellular matrix edit

 
Scanning electron micrograph of mixed-culture biofilm, demonstrating in detail a spatially heterogeneous arrangement of bacterial cells and extracellular polymeric substances.

The EPS matrix consists of exopolysaccharides, proteins and nucleic acids.[42][43][44] A large proportion of the EPS is more or less strongly hydrated, however, hydrophobic EPS also occur; one example is cellulose[45] which is produced by a range of microorganisms. This matrix encases the cells within it and facilitates communication among them through biochemical signals as well as gene exchange. The EPS matrix also traps extracellular enzymes and keeps them in close proximity to the cells. Thus, the matrix represents an external digestion system and allows for stable synergistic microconsortia of different species.[46] Some biofilms have been found to contain water channels that help distribute nutrients and signalling molecules.[47] This matrix is strong enough that under certain conditions, biofilms can become fossilized (stromatolites).

Bacteria living in a biofilm usually have significantly different properties from free-floating bacteria of the same species, as the dense and protected environment of the film allows them to cooperate and interact in various ways.[48] One benefit of this environment is increased resistance to detergents and antibiotics, as the dense extracellular matrix and the outer layer of cells protect the interior of the community.[49][50] In some cases antibiotic resistance can be increased up to 5,000 times.[51] Lateral gene transfer is often facilitated within bacterial and archaeal biofilms[52] and can leads to a more stable biofilm structure.[53] Extracellular DNA is a major structural component of many different microbial biofilms.[54] Enzymatic degradation of extracellular DNA can weaken the biofilm structure and release microbial cells from the surface.

However, biofilms are not always less susceptible to antibiotics. For instance, the biofilm form of Pseudomonas aeruginosa has no greater resistance to antimicrobials than do stationary-phase planktonic cells, although when the biofilm is compared to logarithmic-phase planktonic cells, the biofilm does have greater resistance to antimicrobials. This resistance to antibiotics in both stationary-phase cells and biofilms may be due to the presence of persister cells.[55]

Habitats edit

 
Mats of bacterial biofilm color the hot springs in Yellowstone National Park. The longest raised mat area is about half a meter long.
 
Thermophilic bacteria in the outflow of Mickey Hot Springs, Oregon, approximately 20 mm thick.

Biofilms are ubiquitous in organic life. Nearly every species of microorganism have mechanisms by which they can adhere to surfaces and to each other. Biofilms will form on virtually every non-shedding surface in non-sterile aqueous or humid environments. Biofilms can grow in the most extreme environments: from, for example, the extremely hot, briny waters of hot springs ranging from very acidic to very alkaline, to frozen glaciers.

Biofilms can be found on rocks and pebbles at the bottoms of most streams or rivers and often form on the surfaces of stagnant pools of water. Biofilms are important components of food chains in rivers and streams and are grazed by the aquatic invertebrates upon which many fish feed. Biofilms are found on the surface of and inside plants. They can either contribute to crop disease or, as in the case of nitrogen-fixing rhizobia on root nodules, exist symbiotically with the plant.[56] Examples of crop diseases related to biofilms include citrus canker, Pierce's disease of grapes, and bacterial spot of plants such as peppers and tomatoes.[57]

Percolating filters edit

Percolating filters in sewage treatment works are highly effective removers of pollutants from settled sewage liquor. They work by trickling the liquid over a bed of hard material which is designed to have a very large surface area. A complex biofilm develops on the surface of the medium which absorbs, adsorbs and metabolises the pollutants. The biofilm grows rapidly and when it becomes too thick to retain its grip on the media it washes off and is replaced by newly grown film. The washed off ("sloughed" off) film is settled out of the liquid stream to leave a highly purified effluent.[58]

Slow sand filter edit

Slow sand filters are used in water purification for treating raw water to produce a potable product. They work through the formation of a biofilm called the hypogeal layer or Schmutzdecke in the top few millimetres of the fine sand layer. The Schmutzdecke is formed in the first 10–20 days of operation[59] and consists of bacteria, fungi, protozoa, rotifera and a range of aquatic insect larvae. As an epigeal biofilm ages, more algae tend to develop and larger aquatic organisms may be present including some bryozoa, snails and annelid worms. The surface biofilm is the layer that provides the effective purification in potable water treatment, the underlying sand providing the support medium for this biological treatment layer. As water passes through the hypogeal layer, particles of foreign matter are trapped in the mucilaginous matrix and soluble organic material is adsorbed. The contaminants are metabolised by the bacteria, fungi and protozoa. The water produced from an exemplary slow sand filter is of excellent quality with 90–99% bacterial cell count reduction.[60]

Rhizosphere edit

Plant-beneficial microbes can be categorized as plant growth-promoting rhizobacteria.[61] These plant growth-promoters colonize the roots of plants, and provide a wide range of beneficial functions for their host including nitrogen fixation, pathogen suppression, anti-fungal properties, and the breakdown of organic materials.[62] One of these functions is the defense against pathogenic, soil-borne bacteria and fungi by way of induced systemic resistance (ISR)[63] or induced systemic responses triggered by pathogenic microbes (pathogen-induced systemic acquired resistance).[64]  Plant exudates act as chemical signals for host specific bacteria to colonize.[65] Rhizobacteria colonization steps include attractions, recognition, adherence, colonization, and growth.[62] Bacteria that have been shown to be beneficial and form biofilms include Bacillus, Pseudomonas, and Azospirillum.[66][67]  Biofilms in the rhizosphere often result in pathogen or plant induced systemic resistances.  Molecular properties on the surface of the bacterium cause an immune response in the plant host.[65]  These microbe associated molecules interact with receptors on the surface of plant cells, and activate a biochemical response that is thought to include several different genes at a number of loci.[65] Several other signaling molecules have been linked to both induced systemic responses and pathogen-induced systemic responses, such as jasmonic acid and ethylene.[62]  Cell envelope components such as bacterial flagella and lipopolysaccharides, which are recognized by plant cells as components of pathogens.[68]  Certain iron metabolites produced by Pseudomonas have also been shown to create an induced systemic response.[65]  This function of the biofilm helps plants build stronger resistance to pathogens.

Plants that have been colonized by PGPR forming a biofilm have gained systemic resistances and are primed for defense against pathogens. This means that the genes necessary for the production of proteins that work towards defending the plant against pathogens have been expressed, and the plant has a "stockpile" of compounds to release to fight off pathogens.[65]  A primed defense system is much faster in responding to pathogen induced infection, and may be able to deflect pathogens before they are able to establish themselves.[69] Plants increase the production of lignin, reinforcing cell walls and making it difficult for pathogens to penetrate into the cell, while also cutting off nutrients to already infected cells, effectively halting the invasion.[62]  They produce antimicrobial compounds such as phytoalexins, chitinases, and proteinase inhibitors, which prevent the growth of pathogens.[64]  These functions of disease suppression and pathogen resistance ultimately lead to an increase in agricultural production and a decrease in the use of chemical pesticides, herbicides, and fungicides because there is a reduced amount of crop loss due to disease.[70]  Induced systemic resistance and pathogen-induced systemic acquired resistance are both potential functions of biofilms in the rhizosphere, and should be taken into consideration when applied to new age agricultural practices because of their effect on disease suppression without the use of dangerous chemicals.

Mammalian gut edit

Studies in 2003 discovered that the immune system supports biofilm development in the large intestine. This was supported mainly with the fact that the two most abundantly produced molecules by the immune system also support biofilm production and are associated with the biofilms developed in the gut. This is especially important because the appendix holds a mass amount of these bacterial biofilms.[71] This discovery helps to distinguish the possible function of the appendix and the idea that the appendix can help reinoculate the gut with good gut flora. However, modified or disrupted states of biofilms in the gut have been connected to diseases such as inflammatory bowel disease and colorectal cancer.[72]

Human environment edit

In the human environment, biofilms can grow in showers very easily since they provide a moist and warm environment for them to thrive. They can form inside water and sewage pipes and cause clogging and corrosion. On floors and counters, they can make sanitation difficult in food preparation areas. In soil, they can cause bioclogging. In cooling- or heating-water systems, they are known to reduce heat transfer.[73] Biofilms in marine engineering systems, such as pipelines of the offshore oil and gas industry,[74] can lead to substantial corrosion problems. Corrosion is mainly due to abiotic factors; however, at least 20% of corrosion is caused by microorganisms that are attached to the metal subsurface (i.e., microbially influenced corrosion).

Ship fouling edit

Bacterial adhesion to boat hulls serves as the foundation for biofouling of seagoing vessels. Once a film of bacteria forms, it is easier for other marine organisms such as barnacles to attach. Such fouling can reduce maximum vessel speed by up to 20%, prolonging voyages and consuming fuel. Time in dry dock for refitting and repainting reduces the productivity of shipping assets, and the useful life of ships is also reduced due to corrosion and mechanical removal (scraping) of marine organisms from ships' hulls.

Stromatolites edit

Stromatolites are layered accretionary structures formed in shallow water by the trapping, binding and cementation of sedimentary grains by microbial biofilms, especially of cyanobacteria. Stromatolites include some of the most ancient records of life on Earth, and are still forming today.

Dental plaque edit

Within the human body, biofilms are present on the teeth as dental plaque, where they may cause tooth decay and gum disease. These biofilms can either be in an uncalcified state that can be removed by dental instruments, or a calcified state which is more difficult to remove. Removal techniques can also include antimicrobials.[75]

Dental plaque is an oral biofilm that adheres to the teeth and consists of many species of both bacteria and fungi (such as Streptococcus mutans and Candida albicans), embedded in salivary polymers and microbial extracellular products. The accumulation of microorganisms subjects the teeth and gingival tissues to high concentrations of bacterial metabolites which results in dental disease.[76] Biofilm on the surface of teeth is frequently subject to oxidative stress[77] and acid stress.[78] Dietary carbohydrates can cause a dramatic decrease in pH in oral biofilms to values of 4 and below (acid stress).[78] A pH of 4 at body temperature of 37 °C causes depurination of DNA, leaving apurinic (AP) sites in DNA,[79] especially loss of guanine.[80]

Dental plaque biofilm can result in dental caries if it is allowed to develop over time. An ecologic shift away from balanced populations within the dental biofilm is driven by certain (cariogenic) microbiological populations beginning to dominate when the environment favors them. The shift to an acidogenic, aciduric, and cariogenic microbiological population develops and is maintained by frequent consumption of fermentable dietary carbohydrate. The resulting activity shift in the biofilm (and resulting acid production within the biofilm, at the tooth surface) is associated with an imbalance of demineralization over remineralization, leading to net mineral loss within dental hard tissues (enamel and then dentin), the symptom being a carious lesion, or cavity. By preventing the dental plaque biofilm from maturing or by returning it back to a non-cariogenic state, dental caries can be prevented and arrested.[81][82] This can be achieved through the behavioral step of reducing the supply of fermentable carbohydrates (i.e. sugar intake) and frequent removal of the biofilm (i.e., toothbrushing).[81]

Intercellular communication edit

A peptide pheromone quorum sensing signaling system in S. mutans includes the competence stimulating peptide (CSP) that controls genetic competence.[83][84] Genetic competence is the ability of a cell to take up DNA released by another cell. Competence can lead to genetic transformation, a form of sexual interaction, favored under conditions of high cell density and/or stress where there is maximal opportunity for interaction between the competent cell and the DNA released from nearby donor cells. This system is optimally expressed when S. mutans cells reside in an actively growing biofilm. Biofilm grown S. mutans cells are genetically transformed at a rate 10- to 600-fold higher than S. mutans growing as free-floating planktonic cells suspended in liquid.[83]

When the biofilm, containing S. mutans and related oral streptococci, is subjected to acid stress, the competence regulon is induced, leading to resistance to being killed by acid.[78] As pointed out by Michod et al., transformation in bacterial pathogens likely provides for effective and efficient recombinational repair of DNA damages.[85] It appears that S. mutans can survive the frequent acid stress in oral biofilms, in part, through the recombinational repair provided by competence and transformation.

Predator-prey interactions

Predator-prey interactions between biofilms and bacterivores, such as the soil-dwelling nematode Caenorhabditis elegans, had been extensively studied. Via the production of sticky matrix and formation of aggregates, Yersinia pestis biofilms can prevent feeding by obstructing the mouth of C. elegans.[86] Moreover, Pseudomonas aeruginosa biofilms can impede the slithering motility of C. elegans, termed as 'quagmire phenotype', resulting in trapping of C. elegans within the biofilms and preventing the exploration of nematodes to feed on susceptible biofilms.[87] This significantly reduced the ability of predator to feed and reproduce, thereby promoting the survival of biofilms. Pseudomonas aeruginosa biofilms can also mask their chemical signatures, where they reduced the diffusion of quorum sensing molecules into the environment and prevented the detection of C. elegans.[88]

Taxonomic diversity edit

Many different bacteria form biofilms, including gram-positive (e.g. Bacillus spp, Listeria monocytogenes, Staphylococcus spp, and lactic acid bacteria, including Lactobacillus plantarum and Lactococcus lactis) and gram-negative species (e.g. Escherichia coli, or Pseudomonas aeruginosa).[89] Cyanobacteria also form biofilms in aquatic environments.[90]

Biofilms are formed by bacteria that colonize plants, e.g. Pseudomonas putida, Pseudomonas fluorescens, and related pseudomonads which are common plant-associated bacteria found on leaves, roots, and in the soil, and the majority of their natural isolates form biofilms.[91] Several nitrogen-fixing symbionts of legumes such as Rhizobium leguminosarum and Sinorhizobium meliloti form biofilms on legume roots and other inert surfaces.[91]

Along with bacteria, biofilms are also generated by archaea[52] and by a range of eukaryotic organisms, including fungi e.g. Cryptococcus laurentii[92] and microalgae. Among microalgae, one of the main progenitors of biofilms are diatoms, which colonise both fresh and marine environments worldwide.[93][94]

For other species in disease-associated biofilms and biofilms arising from eukaryotes, see below.

Infectious diseases edit

Biofilms have been found to be involved in a wide variety of microbial infections in the body, by one estimate 80% of all infections.[95] Infectious processes in which biofilms have been implicated include common problems such as bacterial vaginosis, urinary tract infections, catheter infections, middle-ear infections, formation of dental plaque,[96] gingivitis, coating contact lenses,[97] and less common but more lethal processes such as endocarditis, infections in cystic fibrosis, and infections of permanent indwelling devices such as joint prostheses, heart valves, and intervertebral disc.[98][99][100] The first visual evidence of a biofilm was recorded after spine surgery.[101] It was found that in the absence of clinical presentation of infection, impregnated bacteria could form a biofilm around an implant, and this biofilm can remain undetected via contemporary diagnostic methods, including swabbing. Implant biofilm is frequently present in "aseptic" pseudarthrosis cases.[101][102][103] Furthermore, it has been noted that bacterial biofilms may impair cutaneous wound healing and reduce topical antibacterial efficiency in healing or treating infected skin wounds.[104] The diversity of P. aeruginosa cells within a biofilm is thought to make it harder to treat the infected lungs of people with cystic fibrosis.[14] Early detection of biofilms in wounds is crucial to successful chronic wound management. Although many techniques have developed to identify planktonic bacteria in viable wounds, few have been able to quickly and accurately identify bacterial biofilms. Future studies are needed to find means of identifying and monitoring biofilm colonization at the bedside to permit timely initiation of treatment.[105]

It has been shown that biofilms are present on the removed tissue of 80% of patients undergoing surgery for chronic sinusitis. The patients with biofilms were shown to have been denuded of cilia and goblet cells, unlike the controls without biofilms who had normal cilia and goblet cell morphology.[106] Biofilms were also found on samples from two of 10 healthy controls mentioned. The species of bacteria from intraoperative cultures did not correspond to the bacteria species in the biofilm on the respective patient's tissue. In other words, the cultures were negative though the bacteria were present.[107] New staining techniques are being developed to differentiate bacterial cells growing in living animals, e.g. from tissues with allergy-inflammations.[108]

Research has shown that sub-therapeutic levels of β-lactam antibiotics induce biofilm formation in Staphylococcus aureus. This sub-therapeutic level of antibiotic may result from the use of antibiotics as growth promoters in agriculture, or during the normal course of antibiotic therapy. The biofilm formation induced by low-level methicillin was inhibited by DNase, suggesting that the sub-therapeutic levels of antibiotic also induce extracellular DNA release.[109] Moreover, from an evolutionary point of view, the creation of the tragedy of the commons in pathogenic microbes may provide advanced therapeutic ways for chronic infections caused by biofilms via genetically engineered invasive cheaters who can invade wild-types 'cooperators' of pathogenic bacteria until cooperator populations go to extinction or overall population 'cooperators and cheaters ' go to extinction.[110]

Pseudomonas aeruginosa edit

P. aeruginosa represents a commonly used biofilm model organism since it is involved in different types of biofilm-associated chronic infections.[42] Examples of such infections include chronic wounds, chronic otitis media, chronic prostatitis and chronic lung infections in cystic fibrosis (CF) patients. About 80% of CF patients have chronic lung infection, caused mainly by P. aeruginosa growing in a non-surface attached biofilms surround by PMN.[111] The infection remains present despite aggressive antibiotic therapy and is a common cause of death in CF patients due to constant inflammatory damage to the lungs.[42] In patients with CF, one therapy for treating early biofilm development is to employ DNase to structurally weaken the biofilm.[5][112]

Biofilm formation of P. aeruginosa, along with other bacteria, is found in 90% of chronic wound infections, which leads to poor healing and high cost of treatment estimated at more than US$25 billion every year in the United States.[113] In order to minimize the P. aeruginosa infection, host epithelial cells secrete antimicrobial peptides, such as lactoferrin, to prevent the formation of the biofilms.[114]

Streptococcus pneumoniae edit

Streptococcus pneumoniae is the main cause of community-acquired pneumonia and meningitis in children and the elderly, and of sepsis in HIV-infected persons. When S. pneumoniae grows in biofilms, genes are specifically expressed that respond to oxidative stress and induce competence.[115] Formation of a biofilm depends on competence stimulating peptide (CSP). CSP also functions as a quorum-sensing peptide. It not only induces biofilm formation, but also increases virulence in pneumonia and meningitis.

It has been proposed that competence development and biofilm formation is an adaptation of S. pneumoniae to survive the defenses of the host.[85] In particular, the host's polymorphonuclear leukocytes produce an oxidative burst to defend against the invading bacteria, and this response can kill bacteria by damaging their DNA. Competent S. pneumoniae in a biofilm have the survival advantage that they can more easily take up transforming DNA from nearby cells in the biofilm to use for recombinational repair of oxidative damages in their DNA. Competent S. pneumoniae can also secrete an enzyme (murein hydrolase) that destroys non-competent cells (fratricide) causing DNA to be released into the surrounding medium for potential use by the competent cells.[116]

The insect antimicrobial peptide cecropin A can destroy planktonic and sessile biofilm-forming uropathogenic E. coli cells, either alone or when combined with the antibiotic nalidixic acid, synergistically clearing infection in vivo (in the insect host Galleria mellonella) without off-target cytotoxicity. The multi-target mechanism of action involves outer membrane permeabilization followed by biofilm disruption triggered by the inhibition of efflux pump activity and interactions with extracellular and intracellular nucleic acids.[117]

Escherichia coli edit

Escherichia coli biofilms are responsible for many intestinal infectious diseases.[118] The Extraintestinal group of E. coli (ExPEC) is the dominant bacterial group that attacks the urinary system, which leads to urinary tract infections.[119] The biofilm formation of these pathogenic E. coli is hard to eradicate due to the complexity of its aggregation structure, and it has a significant contribution to developing aggressive medical complications, increase in hospitalization rate, and cost of treatment.[120][121] The development of E. coli biofilm is a common leading cause of urinary tract infections (UTI) in hospitals through its contribution to developing medical device-associated infections. Catheter-associated urinary tract infections (CAUTI) represent the most common hospital-acquired infection due to the formation of the pathogenic E. coli biofilm inside the catheters.[122]

Staphylococcus aureus edit

Staphylococcus aureus pathogen can attack skin and lungs, leading to skin infection and pneumonia.[123][124] Moreover, the biofilm infections network of S. aureus plays a critical role in preventing immune cells, such as macrophages from eliminating and destroying bacterial cells.[125] Furthermore, biofilm formation by bacteria, such as S. aureus, not only develops resistance against antibiotic medication but also develop internal resistance toward antimicrobial peptides (AMPs), leading to preventing the inhibition of the pathogen and maintaining its survival.[126]

Uses and impact edit

In medicine edit

It is suggested that around two-thirds of bacterial infections in humans involve biofilms.[51][127] Infections associated with the biofilm growth usually are challenging to eradicate.[128] This is mostly due to the fact that mature biofilms display antimicrobial tolerance, and immune response evasions.[129][42] Biofilms often form on the inert surfaces of implanted devices such as catheters, prosthetic cardiac valves and intrauterine devices.[130] Some of the most difficult infections to treat are those associated with the use of medical devices.[51][102]

The rapidly expanding worldwide industry for biomedical devices and tissue engineering related products is already at $180 billion per year, yet this industry continues to suffer from microbial colonization. No matter the sophistication, microbial infections can develop on all medical devices and tissue engineering constructs.[129] 60-70% of hospital-acquired infections are associated with the implantation of a biomedical device.[129] This leads to 2 million cases annually in the U.S., costing the healthcare system over $5 billion in additional healthcare expenses.[129]

The level of antibiotic resistance in a biofilm is much greater than that of non-biofilm bacteria, and can be as much as 5,000 times greater.[51] The extracellular matrix of biofilm is considered one of the leading factors that can reduce the penetration of antibiotics into a biofilm structure and contributes to antibiotic resistance.[131] Further, it has been demonstrated that the evolution of resistance to antibiotics may be affected by the biofilm lifestyle.[132] Bacteriophage therapy can disperse the biofilm generated by antibiotic-resistant bacteria.[133]

It has been shown that the introduction of a small current of electricity to the liquid surrounding a biofilm, together with small amounts of antibiotic can reduce the level of antibiotic resistance to levels of non-biofilm bacteria. This is termed the bioelectric effect.[51][134] The application of a small DC current on its own can cause a biofilm to detach from its surface.[51] A study showed that the type of current used made no difference to the bioelectric effect.[134]

In industry edit

Biofilms can also be harnessed for constructive purposes. For example, many sewage treatment plants include a secondary treatment stage in which waste water passes over biofilms grown on filters, which extract and digest organic compounds. In such biofilms, bacteria are mainly responsible for removal of organic matter (BOD), while protozoa and rotifers are mainly responsible for removal of suspended solids (SS), including pathogens and other microorganisms. Slow sand filters rely on biofilm development in the same way to filter surface water from lake, spring or river sources for drinking purposes. What is regarded as clean water is effectively a waste material to these microcellular organisms. Biofilms can help eliminate petroleum oil from contaminated oceans or marine systems. The oil is eliminated by the hydrocarbon-degrading activities of communities of hydrocarbonoclastic bacteria (HCB).[135] Biofilms are used in microbial fuel cells (MFCs) to generate electricity from a variety of starting materials, including complex organic waste and renewable biomass.[8][136][137] Biofilms are also relevant for the improvement of metal dissolution in bioleaching industry,[138] and aggregation of microplastics pollutants for convenient removal from the environment.[139][140]

Food industry edit

Biofilms have become problematic in several food industries due to the ability to form on plants and during industrial processes.[141] Bacteria can survive long periods of time in water, animal manure, and soil, causing biofilm formation on plants or in the processing equipment.[142] The buildup of biofilms can affect the heat flow across a surface and increase surface corrosion and frictional resistance of fluids.[143] These can lead to a loss of energy in a system and overall loss of products.[143] Along with economic problems, biofilm formation on food poses a health risk to consumers due to the ability to make the food more resistant to disinfectants[141] As a result, from 1996 to 2010 the Centers for Disease Control and Prevention estimated 48 million foodborne illnesses per year.[141] Biofilms have been connected to about 80% of bacterial infections in the United States.[141]

In produce, microorganisms attach to the surfaces and biofilms develop internally.[141] During the washing process, biofilms resist sanitization and allow bacteria to spread across the produce,[141] especially via kitchen utensils.[144] This problem is also found in ready-to-eat foods, because the foods go through limited cleaning procedures before consumption[141] Due to the perishability of dairy products and limitations in cleaning procedures, resulting in the buildup of bacteria, dairy is susceptible to biofilm formation and contamination.[141][143] The bacteria can spoil the products more readily and contaminated products pose a health risk to consumers. One species of bacteria that can be found in various industries and is a major cause of foodborne disease is Salmonella.[145] Large amounts of Salmonella contamination can be found in the poultry processing industry as about 50% of Salmonella strains can produce biofilms on poultry farms.[141] Salmonella increases the risk of foodborne illnesses when the poultry products are not cleaned and cooked correctly. Salmonella is also found in the seafood industry where biofilms form from seafood borne pathogens on the seafood itself as well as in water.[145] Shrimp products are commonly affected by Salmonella because of unhygienic processing and handling techniques[145] The preparation practices of shrimp and other seafood products can allow for bacteria buildup on the products.[145]

New forms of cleaning procedures are being tested to reduce biofilm formation in these processes which will lead to safer and more productive food processing industries. These new forms of cleaning procedures also have a profound effect on the environment, often releasing toxic gases into the groundwater reservoirs.[143] As a response to the aggressive methods employed in controlling biofilm formation, there are a number of novel technologies and chemicals under investigation that can prevent either the proliferation or adhesion of biofilm-secreting microbes. Latest proposed biomolecules presenting marked anti-biofilm activity include a range of metabolites such as bacterial rhamnolipids[146] and even plant-[147] and animal-derived alkaloids.[148]

In aquaculture edit

 
A biofilm from the Dead Sea

In shellfish and algal aquaculture, biofouling microbial species tend to block nets and cages and ultimately outcompete the farmed species for space and food.[149] Bacterial biofilms start the colonization process by creating microenvironments that are more favorable for biofouling species. In the marine environment, biofilms could reduce the hydrodynamic efficiency of ships and propellers, lead to pipeline blockage and sensor malfunction, and increase the weight of appliances deployed in seawater.[150] Numerous studies have shown that biofilm can be a reservoir for potentially pathogenic bacteria in freshwater aquaculture.[151][152][153][154] Moreover, biofilms are important in establishing infections on the fish.[155] As mentioned previously, biofilms can be difficult to eliminate even when antibiotics or chemicals are used in high doses.[156][157] The role that biofilm plays as reservoirs of bacterial fish pathogens has not been explored in detail but it certainly deserves to be studied.

Eukaryotic edit

Along with bacteria, biofilms are often initiated and produced by eukaryotic microbes. The biofilms produced by eukaryotes is usually occupied by bacteria and other eukaryotes alike, however the surface is cultivated and EPS is secreted initially by the eukaryote.[92][93][158] Both fungi and microalgae are known to form biofilms in such a way. Biofilms of fungal origin are important aspects of human infection and fungal pathogenicity, as the fungal infection is more resistant to antifungals.[159][160]

In the environment, fungal biofilms are an area of ongoing research. One key area of research is fungal biofilms on plants. For example, in the soil, plant associated fungi including mycorrhiza have been shown to decompose organic matter and protect plants from bacterial pathogens.[161]

Biofilms in aquatic environments are often founded by diatoms. The exact purpose of these biofilms is unknown, however there is evidence that the EPS produced by diatoms facilitates both cold and salinity stress.[94][162] These eukaryotes interact with a diverse range of other organisms within a region known as the phycosphere, but importantly are the bacteria associated with diatoms, as it has been shown that although diatoms excrete EPS, they only do so when interacting with certain bacteria species.[163][164]

Horizontal gene transfer edit

Horizontal gene transfer is the lateral transfer of genetic material between cellular organisms. It happens frequently in prokaryotes, and less frequently in eukaryotes. In bacteria, horizontal gene transfer can occur through transformation (uptake of free floating DNA in the environment), transduction (virus mediated DNA uptake), or conjugation (transfer of DNA between pili structures of two adjacent bacteria).[165] Recent studies have also uncovered other mechanisms, such as membrane vesicle transmission or gene transfer agents.[166] Biofilms promote horizontal gene transfer in a variety of ways.

 
Transmission electron micrograph showing bacteria Escherichia coli that form extensive biofilms using a network of conjugative F-pili. Source: Jonasz Patkowski

Bacterial conjugation has been shown to accelerate biofilm formation in difficult environment due to the robust connections established by the conjugative pili.[167] These connections can often foster cross-species transfer events due to the diverse heterogeneity of many biofilms. Additionally, biofilms are structurally confined by a polysaccharide matrix, providing the close spatial requirements for conjugation. Transformation is also frequently observed in biofilms. Bacterial autolysis is a key mechanism in biofilm structural regulation, providing an abundant source of competent DNA primed for transformative uptake.[168][166] In some instances, inter-biofilm quorum sensing can enhance the competence of free floating eDNA, further promoting transformation.[166] Stx gene transfer through bacteriophage carriers has been witnessed within biofilms, which suggests that biofilms are also a suitable environment for transduction.[166] Membrane vesicles HGT occurs when released membrane vesicles (containing genetic information) fuse with a recipient bacteria, and release genetic material into the bacteria's cytoplasm.[166] Recent research has revealed that membrane vesicle HGT can promote single-strain biofilm formation, yet the role membrane vesicle HGT plays in the formation of multistrain biofilms is still unknown.[166] GTAs, or gene transfer agents, are phage-like particles produced by the host bacteria and contain random DNA fragments from the host bacteria genome.[166] HGT within biofilms can confer antibiotic resistance or increased pathogenicity across the biofilms' population, promoting biofilm homeostasis.[166]

Examples edit

Conjugative plasmids may encode biofilm-associated proteins, such as PtgA, PrgB, or PrgC which promote cell adhesion (required for early biofilm formation).[169] Genes encoding type III fimbriae are found in pOLA52 (Klebsiella pneumoniae plasmid) which promote conjugative-pilus-dependent biofilm formation.[169]

Transformation commonly occurs within biofilms. A phenomenon called fratricide can be seen among streptococcal species in which cell-wall degrading enzymes are released, lysing neighboring bacteria and releasing their DNA. This DNA can then be taken up by the surviving bacteria (transformation).[169] Competence stimulating peptides may play an important role in biofilm formation among S. pneumoniae and S. mutans as well.[169] Among V. cholerae, the competence pilus itself promotes cell aggregation through pilus-pilus interactions at the beginning of biofilm formation.[169]

Phage invasion may play a role in biofilm life cycles, lysing bacteria and releasing their eDNA, which strengthens biofilm structures and can be taken up by neighboring bacteria in transformation.[169] Biofilm destruction caused by the E. coli phage Rac and the P. aeruginosa prophage Pf4 causes detachment of cells from the biofilm.[169] Detachment is a biofilm phenomenon which requires more study, but is hypothesized to proliferate the bacterial species that comprise the biofilm.

Membrane vesicle HGT has been witnessed occurring in marine environments, among Neisseria gonorrhoeae, Pseudomonas aeruginosa, Helicobacter pylori, and among many other bacterial species.[169] Even though membrane vesicle HGT has been shown as a contributing factor in biofilm formation, research is still required to prove that membrane vesicle mediated HGT occurs within biofilms.[166][169] Membrane vesicle HGT has also been shown to modulate phage-bacteria interactions in Bacillus subtilis SPP1 phage-resistant cells (lacking the SPP1 receptor protein). Upon exposure to vesicles containing receptors, transduction of pBT163 (a cat-encoding plasmid) occurs, resulting in the expression of the SPP1 receptor protein, opening the receptive bacteria to future phage infection.[169]

Recent research has shown that the archaeal species H. volcanii has some biofilm phenotypes similar to bacterial biofilms such as differentiation and HGT, which required cell-cell contact and involved formation of cytosolic bridges and cellular fusion events.[170]

Cultivation devices edit

There is a wide variety of biofilm cultivation devices to mimic natural or industrial environments. Although it is important to consider that the particular experimental platform for biofilm research determines what kind of biofilm is cultivated and the data that can be extracted. These devices can be grouped into the following:[171]

  • microtiter plate (MTP) systems and MBEC Assay® [formerly the Calgary Biofilm Device (CBD)]
  • BioFilm Ring Test 26 October 2021 at the Wayback Machine (BRT) or clinical Biofilm Ring Test (cBRT)
  • Robbins Device or modified Robbins Device (such as the MPMR-10PMMA or the Bio-inLine Biofilm Reactor)
  • Drip Flow Biofilm Reactor®
  • rotary devices (such as the CDC Biofilm Reactor®, the Rotating Disk Reactor, the Biofilm Annular Reactor, the Industrial Surfaces Biofilm Reactor, or the Constant Depth Film Fermenter)
  • flow chambers or flow cells (such as the Coupon Evaluation Flow Cell, Transmission Flow Cell, and Capillary Flow Cell from BioSurface Technologies)
  • microfluidic approaches, such as 3D-bacterial “biofilm-dispersal-then-recolonization” (BDR) microfluidic model [40]

See also edit

  • Amyloid – Insoluble protein aggregate with a fibrillar morphology
  • Bacterial nanowires – Electrically conductive appendages produced by a number of bacteria
  • Biofilm factory – The use of microbial biofilms for chemical production
  • Biofilm prevention – Stopping microbes attaching to a surface
  • Biomineralization – Process by which living organisms produce minerals
  • Center for Biofilm Engineering – interdisciplinary research, education, and technology transfer institute of Montana State University
  • Curli – A proteinaceous extracellular fiber produced by enteric bacteria
  • Floc (biofilm) – Type of microbial aggregate suspension
  • Microbial intelligence – Adaptive behavior by microscopic organisms including bacteria and protists
  • Microbial mat – Multi-layered sheet of microorganisms
  • Phage therapy – Therapeutic use of bacteriophages to treat bacterial infections
  • Phototrophic biofilm – Microbial communities including microorganisms which use light as their energy source
  • Stromatolite – Layered sedimentary structure formed by the growth of bacteria or algae

References edit

  1. ^ Vert M, Doi Y, Hellwich KH, Hess M, Hodge P, Kubisa P, Rinaudo M, Schué F (2012). "Terminology for biorelated polymers and applications (IUPAC Recommendations 2012)". Pure and Applied Chemistry. 84 (2): 377–410. doi:10.1351/PAC-REC-10-12-04.
  2. ^ "Biofilms - an overview | ScienceDirect Topics". www.sciencedirect.com. Retrieved 28 January 2024.
  3. ^ a b c López D, Vlamakis H, Kolter R (July 2010). "Biofilms". Cold Spring Harbor Perspectives in Biology. 2 (7): a000398. doi:10.1101/cshperspect.a000398. PMC 2890205. PMID 20519345.
  4. ^ a b c d e Hall-Stoodley L, Costerton JW, Stoodley P (February 2004). "Bacterial biofilms: from the natural environment to infectious diseases". Nature Reviews. Microbiology. 2 (2): 95–108. doi:10.1038/nrmicro821. PMID 15040259. S2CID 9107205.
  5. ^ a b Aggarwal S, Stewart PS, Hozalski RM (January 2016). "Biofilm Cohesive Strength as a Basis for Biofilm Recalcitrance: Are Bacterial Biofilms Overdesigned?". Microbiology Insights. 8 (Suppl 2): 29–32. doi:10.4137/MBI.S31444. PMC 4718087. PMID 26819559.
  6. ^ a b Watnick P, Kolter R (May 2000). "Biofilm, city of microbes". Journal of Bacteriology. 182 (10): 2675–9. doi:10.1128/jb.182.10.2675-2679.2000. PMC 101960. PMID 10781532.
  7. ^ "Building Codes for Bacterial Cities | Quanta Magazine". Quanta Magazine. Retrieved 25 July 2017.
  8. ^ a b Lear G, Lewis GD, eds. (2012). Microbial Biofilms: Current Research and Applications. Caister Academic Press. ISBN 978-1-904455-96-7.
  9. ^ a b O'Toole GA, Kolter R (May 1998). "Initiation of biofilm formation in Pseudomonas fluorescens WCS365 proceeds via multiple, convergent signalling pathways: a genetic analysis". Molecular Microbiology. 28 (3): 449–61. doi:10.1046/j.1365-2958.1998.00797.x. PMID 9632250. S2CID 43897816.
  10. ^ O'Toole GA, Kolter R (October 1998). "Flagellar and twitching motility are necessary for Pseudomonas aeruginosa biofilm development". Molecular Microbiology. 30 (2): 295–304. doi:10.1046/j.1365-2958.1998.01062.x. PMID 9791175. S2CID 25140899.
  11. ^ Karatan E, Watnick P (June 2009). "Signals, regulatory networks, and materials that build and break bacterial biofilms". Microbiology and Molecular Biology Reviews. 73 (2): 310–47. doi:10.1128/MMBR.00041-08. PMC 2698413. PMID 19487730.
  12. ^ Hoffman LR, D'Argenio DA, MacCoss MJ, Zhang Z, Jones RA, Miller SI (August 2005). "Aminoglycoside antibiotics induce bacterial biofilm formation". Nature. 436 (7054): 1171–5. Bibcode:2005Natur.436.1171H. doi:10.1038/nature03912. PMID 16121184. S2CID 4404961. (primary source)
  13. ^ An D, Parsek MR (June 2007). "The promise and peril of transcriptional profiling in biofilm communities". Current Opinion in Microbiology. 10 (3): 292–6. doi:10.1016/j.mib.2007.05.011. PMID 17573234.
  14. ^ a b Momeni B (June 2018). "Division of Labor: How Microbes Split Their Responsibility". Current Biology. 28 (12): R697–R699. doi:10.1016/j.cub.2018.05.024. PMID 29920261. S2CID 49315067.
  15. ^ Case C, Funke B, Tortora G. Microbiology An Introduction (tenth ed.).
  16. ^ Briandet R, Herry J, Bellon-Fontaine M (August 2001). "Determination of the van der Waals, electron donor and electron acceptor surface tension components of static Gram-positive microbial biofilms". Colloids Surf B Biointerfaces. 21 (4): 299–310. doi:10.1016/S0927-7765(00)00213-7. PMID 11397632.
  17. ^ Takahashi H, Suda T, Tanaka Y, Kimura B (June 2010). "Cellular hydrophobicity of Listeria monocytogenes involves initial attachment and biofilm formation on the surface of polyvinyl chloride". Lett. Appl. Microbiol. 50 (6): 618–25. doi:10.1111/j.1472-765X.2010.02842.x. PMID 20438621. S2CID 24880220.
  18. ^ "7: Archaea". Biology LibreTexts. 6 February 2018.
  19. ^ Madigan M (2019). Brock biology of microorganisms (Fifteenth, Global ed.). Pearson. p. 86. ISBN 9781292235103.
  20. ^ a b Wang F, Cvirkaite-Krupovic V, Krupovic M, Egelman EH (June 2022). "Archaeal bundling pili of Pyrobaculum calidifontis reveal similarities between archaeal and bacterial biofilms". Proceedings of the National Academy of Sciences of the United States of America. 119 (26): e2207037119. Bibcode:2022PNAS..11907037W. doi:10.1073/pnas.2207037119. PMC 9245690. PMID 35727984.
  21. ^ "Golden Dome Cave". National Park Service. November 6, 2021. Retrieved February 11, 2024.
  22. ^ a b c Donlan RM (2002). "Biofilms: Microbial Life on Surfaces". Emerging Infectious Diseases. 8 (9): 881–890. doi:10.3201/eid0809.020063. PMC 2732559. PMID 12194761.
  23. ^ Li S, Liu SY, Chan SY, Chua SL (January 2022). "Biofilm matrix cloaks bacterial quorum sensing chemoattractants from predator detection". The ISME Journal. 16 (5): 1388–1396. doi:10.1038/s41396-022-01190-2. PMC 9038794. PMID 35034106.
  24. ^ Ciofu O, Tolker-Nielsen T (2019). "Tolerance and Resistance of Pseudomonas aeruginosa Biofilms to Antimicrobial Agents-How P. aeruginosa Can Escape Antibiotics". Frontiers in Microbiology. 10: 913. doi:10.3389/fmicb.2019.00913. PMC 6509751. PMID 31130925.
  25. ^ Sakuragi Y, Kolter R (July 2007). "Quorum-sensing regulation of the biofilm matrix genes (pel) of Pseudomonas aeruginosa". Journal of Bacteriology. 189 (14): 5383–6. doi:10.1128/JB.00137-07. PMC 1951888. PMID 17496081.
  26. ^ a b Rapacka-Zdonczyk A, Wozniak A, Nakonieczna J, Grinholc M (February 2021). "Development of Antimicrobial Phototreatment Tolerance: Why the Methodology Matters". International Journal of Molecular Sciences. MDPI AG. 22 (4): 2224. doi:10.3390/ijms22042224. PMC 7926562. PMID 33672375.   Material was copied from this source, which is available under a Creative Commons Attribution 4.0 International License.
  27. ^ Hall CW, Mah TF (May 2017). "Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria". FEMS Microbiology Reviews. Oxford University Press (OUP). 41 (3): 276–301. doi:10.1093/femsre/fux010. PMID 28369412.
  28. ^ O'Toole G, Kaplan HB, Kolter R (2000). "Biofilm formation as microbial development". Annual Review of Microbiology. 54: 49–79. doi:10.1146/annurev.micro.54.1.49. PMID 11018124.
  29. ^ a b Monroe D (November 2007). "Looking for chinks in the armor of bacterial biofilms". PLOS Biology. 5 (11): e307. doi:10.1371/journal.pbio.0050307. PMC 2071939. PMID 18001153.
  30. ^ Kaplan JB, Ragunath C, Ramasubbu N, Fine DH (August 2003). "Detachment of Actinobacillus actinomycetemcomitans biofilm cells by an endogenous beta-hexosaminidase activity". Journal of Bacteriology. 185 (16): 4693–8. doi:10.1128/JB.185.16.4693-4698.2003. PMC 166467. PMID 12896987.
  31. ^ Izano EA, Amarante MA, Kher WB, Kaplan JB (January 2008). "Differential roles of poly-N-acetylglucosamine surface polysaccharide and extracellular DNA in Staphylococcus aureus and Staphylococcus epidermidis biofilms". Applied and Environmental Microbiology. 74 (2): 470–6. Bibcode:2008ApEnM..74..470I. doi:10.1128/AEM.02073-07. PMC 2223269. PMID 18039822.
  32. ^ Kaplan JB, Ragunath C, Velliyagounder K, Fine DH, Ramasubbu N (July 2004). "Enzymatic detachment of Staphylococcus epidermidis biofilms". Antimicrobial Agents and Chemotherapy. 48 (7): 2633–6. doi:10.1128/AAC.48.7.2633-2636.2004. PMC 434209. PMID 15215120.
  33. ^ Xavier JB, Picioreanu C, Rani SA, van Loosdrecht MC, Stewart PS (December 2005). "Biofilm-control strategies based on enzymic disruption of the extracellular polymeric substance matrix--a modelling study". Microbiology. 151 (Pt 12): 3817–32. doi:10.1099/mic.0.28165-0. PMID 16339929.
  34. ^ Davies DG, Marques CN (March 2009). "A fatty acid messenger is responsible for inducing dispersion in microbial biofilms". Journal of Bacteriology. 191 (5): 1393–403. doi:10.1128/JB.01214-08. PMC 2648214. PMID 19074399.
  35. ^ Barraud N, Hassett DJ, Hwang SH, Rice SA, Kjelleberg S, Webb JS (2006). "Involvement of nitric oxide in biofilm dispersal of Pseudomonas aeruginosa". Journal of Bacteriology. 188 (21): 7344–7353. doi:10.1128/jb.00779-06. PMC 1636254. PMID 17050922.
  36. ^ Barraud N, Storey MV, Moore ZP, Webb JS, Rice SA, Kjelleberg S (2009). "Nitric oxide-mediated dispersal in single- and multi-species biofilms of clinically and industrially relevant microorganisms". Microbial Biotechnology. 2 (3): 370–378. doi:10.1111/j.1751-7915.2009.00098.x. PMC 3815757. PMID 21261931.
  37. ^ "Dispersal of Biofilm in Cystic Fibrosis using Low Dose Nitric Oxide". University of Southampton. Retrieved 20 January 2012.
  38. ^ a b Chua SL, Liu Y, Yam JK, Tolker-Nielsen T, Kjelleberg S, Givskov M, Yang L (2014). "Dispersed cells represent a distinct stage in the transition from bacterial biofilm to planktonic lifestyles". Nature Communications. 5: 4462. Bibcode:2014NatCo...5.4462C. doi:10.1038/ncomms5462. PMID 25042103.
  39. ^ Chua SL, Hultqvist LD, Yuan M, Rybtke M, Nielsen TE, Givskov M, Tolker-Nielsen T, Yang L (August 2015). "In vitro and in vivo generation and characterization of Pseudomonas aeruginosa biofilm-dispersed cells via c-di-GMP manipulation". Nat Protoc. 10 (8): 1165–80. doi:10.1038/nprot.2015.067. hdl:10356/84100. PMID 26158442. S2CID 20235088.
  40. ^ a b Ma Y, Deng Y, Hua H, Khoo BL, Chua SL (August 2023). "Distinct bacterial population dynamics and disease dissemination after biofilm dispersal and disassembly". The ISME Journal. 17 (8): 1290–1302. doi:10.1038/s41396-023-01446-5. PMC 10356768. PMID 37270584.
  41. ^ Nadell CD, Xavier JB, Foster KR (January 2009). "The sociobiology of biofilms". FEMS Microbiology Reviews. 33 (1): 206–24. doi:10.1111/j.1574-6976.2008.00150.x. PMID 19067751.
  42. ^ a b c d Rybtke M, Hultqvist LD, Givskov M, Tolker-Nielsen T (November 2015). "Pseudomonas aeruginosa Biofilm Infections: Community Structure, Antimicrobial Tolerance and Immune Response". Journal of Molecular Biology. 427 (23): 3628–45. doi:10.1016/j.jmb.2015.08.016. PMID 26319792.
  43. ^ Danese PN, Pratt LA, Kolter R (June 2000). "Exopolysaccharide production is required for development of Escherichia coli K-12 biofilm architecture". Journal of Bacteriology. 182 (12): 3593–6. doi:10.1128/jb.182.12.3593-3596.2000. PMC 101973. PMID 10852895.
  44. ^ Branda SS, Chu F, Kearns DB, Losick R, Kolter R (February 2006). "A major protein component of the Bacillus subtilis biofilm matrix". Molecular Microbiology. 59 (4): 1229–38. doi:10.1111/j.1365-2958.2005.05020.x. PMID 16430696. S2CID 3041295.
  45. ^ Choong FX, Bäck M, Fahlén S, Johansson LB, Melican K, Rhen M, et al. (23 November 2016). "Salmonella biofilms using luminescent oligothiophenes". npj Biofilms and Microbiomes. 2: 16024. doi:10.1038/npjbiofilms.2016.24. PMC 5515270. PMID 28721253.
  46. ^ Flemming HC, Wingender J, Szewzyk U, Steinberg P, Rice SA, Kjelleberg S (August 2016). "Biofilms: an emergent form of bacterial life". Nature Reviews. Microbiology. 14 (9): 563–75. doi:10.1038/nrmicro.2016.94. PMID 27510863. S2CID 4384131.
  47. ^ Stoodley P, Debeer D, Lewandowski Z (August 1994). "Liquid flow in biofilm systems". Applied and Environmental Microbiology. 60 (8): 2711–6. Bibcode:1994ApEnM..60.2711S. doi:10.1128/aem.60.8.2711-2716.1994. PMC 201713. PMID 16349345.
  48. ^ Vlamakis H, Aguilar C, Losick R, Kolter R (April 2008). "Control of cell fate by the formation of an architecturally complex bacterial community". Genes & Development. 22 (7): 945–53. doi:10.1101/gad.1645008. PMC 2279205. PMID 18381896.
  49. ^ Stewart PS, Costerton JW (July 2001). "Antibiotic resistance of bacteria in biofilms". Lancet. 358 (9276): 135–8. doi:10.1016/S0140-6736(01)05321-1. PMID 11463434. S2CID 46125592.
  50. ^ Pandey R, Mishra SK, Shrestha A (2021). "Characterisation of ESKAPE Pathogens with Special Reference to Multidrug Resistance and Biofilm Production in a Nepalese Hospital". Infect Drug Resist. 14: 2201–2212. doi:10.2147/IDR.S306688. PMC 8214009. PMID 34163185.
  51. ^ a b c d e f Del Pozo JL, Rouse MS, Patel R (September 2008). "Bioelectric effect and bacterial biofilms. A systematic review". The International Journal of Artificial Organs. 31 (9): 786–795. doi:10.1177/039139880803100906. PMC 3910516. PMID 18924090.
  52. ^ a b Chimileski S, Franklin MJ, Papke RT (August 2014). "Biofilms formed by the archaeon Haloferax volcanii exhibit cellular differentiation and social motility, and facilitate horizontal gene transfer". BMC Biology. 12: 65. doi:10.1186/s12915-014-0065-5. PMC 4180959. PMID 25124934.
  53. ^ Molin S, Tolker-Nielsen T (June 2003). "Gene transfer occurs with enhanced efficiency in biofilms and induces enhanced stabilisation of the biofilm structure". Current Opinion in Biotechnology. 14 (3): 255–61. doi:10.1016/S0958-1669(03)00036-3. PMID 12849777.
  54. ^ Jakubovics NS, Shields RC, Rajarajan N, Burgess JG (December 2013). "Life after death: the critical role of extracellular DNA in microbial biofilms". Letters in Applied Microbiology. 57 (6): 467–75. doi:10.1111/lam.12134. PMID 23848166. S2CID 206168952.
  55. ^ Spoering AL, Lewis K (December 2001). "Biofilms and planktonic cells of Pseudomonas aeruginosa have similar resistance to killing by antimicrobials". Journal of Bacteriology. 183 (23): 6746–51. doi:10.1128/JB.183.23.6746-6751.2001. PMC 95513. PMID 11698361.
  56. ^ . Archived from the original on 22 June 2008. (primary source)
  57. ^ Andersen PC, Brodbeck BV, Oden S, Shriner A, Leite B (September 2007). "Influence of xylem fluid chemistry on planktonic growth, biofilm formation and aggregation of Xylella fastidiosa". FEMS Microbiology Letters. 274 (2): 210–7. doi:10.1111/j.1574-6968.2007.00827.x. PMID 17610515.
  58. ^ . Staffordshire University. Archived from the original on 18 April 2011. Retrieved 13 December 2019.
  59. ^ Centre for Affordable Water and Sanitation Technology, Biosand Filter Manual: Design, Construction, & Installation," July 2007.
  60. ^ (PDF). Tech Brief. Morgantown, WV: National Drinking Water Clearinghouse (U.S.). 14. June 2000. Archived from the original (PDF) on 6 April 2016.
  61. ^ Kloepper JW (1988). "Plant Growth-Promoting Rhizobacteria on Canola (Rapeseed)". Plant Disease. 72 (1): 42. doi:10.1094/pd-72-0042. ISSN 0191-2917.
  62. ^ a b c d Nihorimbere V, Cawoy H, Seyer A, Brunelle A, Thonart P, Ongena M (January 2012). "Impact of rhizosphere factors on cyclic lipopeptide signature from the plant beneficial strain Bacillus amyloliquefaciens S499". FEMS Microbiology Ecology. 79 (1): 176–91. Bibcode:2012FEMME..79..176N. doi:10.1111/j.1574-6941.2011.01208.x. PMID 22029651.
  63. ^ Choudhary DK, Johri BN (September 2009). "Interactions of Bacillus spp. and plants—with special reference to induced systemic resistance (ISR)". Microbiological Research. 164 (5): 493–513. doi:10.1016/j.micres.2008.08.007. PMID 18845426.
  64. ^ a b van Loon LC (5 June 2007). "Plant responses to plant growth-promoting rhizobacteria". European Journal of Plant Pathology. 119 (3): 243–254. doi:10.1007/s10658-007-9165-1. ISSN 0929-1873.
  65. ^ a b c d e Van Wees SC, Van der Ent S, Pieterse CM (August 2008). "Plant immune responses triggered by beneficial microbes". Current Opinion in Plant Biology. 11 (4): 443–8. doi:10.1016/j.pbi.2008.05.005. hdl:1874/30010. PMID 18585955. S2CID 25880745.
  66. ^ Holguin G, Bashan Y (December 1996). "Nitrogen-fixation by Azospirillum brasilense Cd is promoted when co-cultured with a mangrove rhizosphere bacterium (Staphylococcus sp.)". Soil Biology and Biochemistry. 28 (12): 1651–1660. doi:10.1016/s0038-0717(96)00251-9. ISSN 0038-0717.
  67. ^ Babalola OO (November 2010). "Beneficial bacteria of agricultural importance". Biotechnology Letters. 32 (11): 1559–70. doi:10.1007/s10529-010-0347-0. PMID 20635120. S2CID 13518392.
  68. ^ Bakker PA, Pieterse CM, van Loon LC (February 2007). "Induced Systemic Resistance by Fluorescent Pseudomonas spp". Phytopathology. 97 (2): 239–43. doi:10.1094/phyto-97-2-0239. PMID 18944381.
  69. ^ Bent E (2006). "Induced Systemic Resistance Mediated by Plant Growth-Promoting Rhizobacteria (PGPR) and Fungi (PGPF)". Multigenic and Induced Systemic Resistance in Plants. Springer US. pp. 225–258. doi:10.1007/0-387-23266-4_10. ISBN 9780387232652.
  70. ^ Lynch JM, Brimecombe MJ, De Leij FA (21 August 2001), "Rhizosphere", eLS, John Wiley & Sons, Ltd, doi:10.1038/npg.els.0000403, ISBN 0470016175
  71. ^ Randal Bollinger R, Barbas AS, Bush EL, Lin SS, Parker W (December 2007). "Biofilms in the large bowel suggest an apparent function of the human vermiform appendix". Journal of Theoretical Biology. 249 (4): 826–31. Bibcode:2007JThBi.249..826R. doi:10.1016/j.jtbi.2007.08.032. PMID 17936308.
  72. ^ Buret AG, Motta JP, Allain T, Ferraz J, Wallace JL (January 2019). "Pathobiont release from dysbiotic gut microbiota biofilms in intestinal inflammatory diseases: a role for iron?". Journal of Biomedical Science. 26 (1): 1. doi:10.1186/s12929-018-0495-4. PMC 6317250. PMID 30602371.
  73. ^ Characklis WG, Nevimons MJ, Picologlou BF (1981). "Influence of Fouling Biofilms on Heat Transfer". Heat Transfer Engineering. 3 (1): 23–37. Bibcode:1981HTrEn...3...23C. doi:10.1080/01457638108939572.
  74. ^ Schwermer CU, Lavik G, Abed RM, Dunsmore B, Ferdelman TG, Stoodley P, et al. (May 2008). "Impact of nitrate on the structure and function of bacterial biofilm communities in pipelines used for injection of seawater into oil fields". Applied and Environmental Microbiology. 74 (9): 2841–51. Bibcode:2008ApEnM..74.2841S. doi:10.1128/AEM.02027-07. PMC 2394879. PMID 18344353.
  75. ^ Chandki R, Banthia P, Banthia R (April 2011). "Biofilms: A microbial home". Journal of Indian Society of Periodontology. 15 (2): 111–4. doi:10.4103/0972-124X.84377. PMC 3183659. PMID 21976832.
  76. ^ Augustin M, Chifiriuc CB, Lazăr V, Stănescu R, Burlibașa M, Ispas DC (December 2010). "Microbial biofilms in dental medicine in reference to implanto-prostethic rehabilitation". Revista de Chirurgie Oro-maxilo-facială și Implantologie (in Romanian). 1 (1): 9–13. ISSN 2069-3850. 8. Retrieved 3 June 2012.[permanent dead link](webpage has a translation button)
  77. ^ Marquis RE (September 1995). "Oxygen metabolism, oxidative stress and acid-base physiology of dental plaque biofilms". Journal of Industrial Microbiology. 15 (3): 198–207. doi:10.1007/bf01569826. PMID 8519478. S2CID 19959528.
  78. ^ a b c Lemos JA, Abranches J, Burne RA (January 2005). (PDF). Current Issues in Molecular Biology. 7 (1): 95–107. PMID 15580782. Archived from the original (PDF) on 7 April 2014. Retrieved 3 April 2014.
  79. ^ Tamm C, Hodes ME, Chargaff E (March 1952). "The formation apurinic acid from the desoxyribonucleic acid of calf thymus". The Journal of Biological Chemistry. 195 (1): 49–63. doi:10.1016/S0021-9258(19)50874-2. PMID 14938354.
  80. ^ Freese EB (April 1961). "Transitions and transversions induced by depurinating agents". Proceedings of the National Academy of Sciences of the United States of America. 47 (4): 540–5. Bibcode:1961PNAS...47..540B. doi:10.1073/pnas.47.4.540. PMC 221484. PMID 13701660.
  81. ^ a b Pennwell, "Toothbrush technology, dentifrices and dental biofilm removal." Dental Academy of CE Accessed 12 January 2022
  82. ^ Fejerskov O (2015). Pathology of dental caries. In: Dental caries: the disease and its clinical management. Oxford (UK): Wiley Blackwell. pp. 7–9. ISBN 978-1405138895.
  83. ^ a b Li YH, Lau PC, Lee JH, Ellen RP, Cvitkovitch DG (February 2001). "Natural genetic transformation of Streptococcus mutans growing in biofilms". J. Bacteriol. 183 (3): 897–908. doi:10.1128/JB.183.3.897-908.2001. PMC 94956. PMID 11208787.
  84. ^ Senadheera D, Cvitkovitch DG (2008). "Quorum Sensing and Biofilm Formation by Streptococcus mutans". Bacterial Signal Transduction: Networks and Drug Targets. Advances in Experimental Medicine and Biology. Vol. 631. pp. 178–88. doi:10.1007/978-0-387-78885-2_12. ISBN 978-0-387-78884-5. PMID 18792689.
  85. ^ a b Michod RE, Bernstein H, Nedelcu AM (May 2008). "Adaptive value of sex in microbial pathogens". Infect. Genet. Evol. 8 (3): 267–85. doi:10.1016/j.meegid.2008.01.002. PMID 18295550.http://www.hummingbirds.arizona.edu/Faculty/Michod/Downloads/IGE%20review%20sex.pdf
  86. ^ Atkinson S, Goldstone RJ, Joshua GW, Chang CY, Patrick HL, Cámara M, et al. (January 2011). "Biofilm development on Caenorhabditis elegans by Yersinia is facilitated by quorum sensing-dependent repression of type III secretion". PLOS Pathogens. 7 (1): e1001250. doi:10.1371/journal.ppat.1001250. PMC 3017118. PMID 21253572.
  87. ^ Chan SY, Liu SY, Seng Z, Chua SL (September 2020). "Biofilm matrix disrupts nematode motility and predatory behavior". The ISME Journal. 15 (1): 260–269. doi:10.1038/s41396-020-00779-9. PMC 7852553. PMID 32958848.
  88. ^ Li S, Liu SY, Chan SY, Chua SL (May 2022). "Biofilm matrix cloaks bacterial quorum sensing chemoattractants from predator detection". The ISME Journal. 16 (5): 1388–1396. doi:10.1038/s41396-022-01190-2. PMC 9038794. PMID 35034106.
  89. ^ Abee T, Kovács AT, Kuipers OP, van der Veen S (April 2011). "Biofilm formation and dispersal in Gram-positive bacteria" (PDF). Current Opinion in Biotechnology. 22 (2): 172–9. doi:10.1016/j.copbio.2010.10.016. hdl:11370/999da2a4-d509-471b-bab5-085dac6ff681. PMID 21109420. S2CID 22024410.
  90. ^ Rossi F, De Philippis R (April 2015). "Role of cyanobacterial exopolysaccharides in phototrophic biofilms and in complex microbial mats". Life. 5 (2): 1218–38. Bibcode:2015Life....5.1218R. doi:10.3390/life5021218. PMC 4500136. PMID 25837843.
  91. ^ a b Danhorn T, Fuqua C (2007). "Biofilm formation by plant-associated bacteria". Annual Review of Microbiology. 61: 401–22. doi:10.1146/annurev.micro.61.080706.093316. PMID 17506679.
  92. ^ a b Joubert LM, Wolfaardt GM, Botha A (August 2006). "Microbial exopolymers link predator and prey in a model yeast biofilm system". Microb. Ecol. 52 (2): 187–97. doi:10.1007/s00248-006-9063-7. PMID 16897306. S2CID 20431229.
  93. ^ a b Van Colen C, Underwood GC, Serôdio J, Paterson DM (2014). "Ecology of intertidal microbial biofilms: Mechanisms, patterns and future research needs". Journal of Sea Research. 92: 2–5. Bibcode:2014JSR....92....2V. doi:10.1016/j.seares.2014.07.003.
  94. ^ a b Aslam SN, Cresswell-Maynard T, Thomas DN, Underwood GJ (December 2012). "Production and Characterization of the Intra- and Extracellular Carbohydrates and Polymeric Substances (EPS) of Three Sea-Ice Diatom Species, and Evidence for a Cryoprotective Role for EPS". J. Phycol. 48 (6): 1494–509. doi:10.1111/jpy.12004. PMID 27009999. S2CID 9226690.
  95. ^ "Research on microbial biofilms (PA-03-047)". NIH, National Heart, Lung, and Blood Institute. 20 December 2002.
  96. ^ Rogers A (2008). Molecular Oral Microbiology. Caister Academic Press. pp. 88–91. ISBN 978-1-904455-24-0.
  97. ^ Imamura Y, Chandra J, Mukherjee PK, Lattif AA, Szczotka-Flynn LB, Pearlman E, et al. (January 2008). "Fusarium and Candida albicans biofilms on soft contact lenses: model development, influence of lens type, and susceptibility to lens care solutions". Antimicrobial Agents and Chemotherapy. 52 (1): 171–82. doi:10.1128/AAC.00387-07. PMC 2223913. PMID 17999966.
  98. ^ Capoor MN, Ruzicka F, Schmitz JE, James GA, Machackova T, Jancalek R, et al. (3 April 2017). "Propionibacterium acnes biofilm is present in intervertebral discs of patients undergoing microdiscectomy". PLOS ONE. 12 (4): e0174518. Bibcode:2017PLoSO..1274518C. doi:10.1371/journal.pone.0174518. PMC 5378350. PMID 28369127.
  99. ^ Lewis K (April 2001). "Riddle of biofilm resistance". Antimicrobial Agents and Chemotherapy. 45 (4): 999–1007. doi:10.1128/AAC.45.4.999-1007.2001. PMC 90417. PMID 11257008.
  100. ^ Parsek MR, Singh PK (2003). "Bacterial biofilms: an emerging link to disease pathogenesis". Annual Review of Microbiology. 57: 677–701. doi:10.1146/annurev.micro.57.030502.090720. PMID 14527295.
  101. ^ a b Agarwal A, Mooney M, Agarwal AG, Jayaswal D, Saakyan G, Goel V, et al. (2020). "High Prevalence of Biofilms on Retrieved Implants from Aseptic Pseudarthrosis Cases". Spine Surgery and Related Research. 5 (2): 104–108. doi:10.22603/ssrr.2020-0147. PMC 8026210. PMID 33842718.
  102. ^ a b Curran N (20 November 2020). "New study first to visually capture biofilm architecture in retrieved implants from live patients". Spinal News International. Retrieved 24 November 2020.
  103. ^ "biofilm". 22 December 2020.
  104. ^ Davis SC, Ricotti C, Cazzaniga A, Welsh E, Eaglstein WH, Mertz PM (2008). "Microscopic and physiologic evidence for biofilm-associated wound colonization in vivo". Wound Repair and Regeneration. 16 (1): 23–9. doi:10.1111/j.1524-475X.2007.00303.x. PMID 18211576. S2CID 205669081.
  105. ^ Vyas KS, Wong LK (January 2016). "Detection of Biofilm in Wounds as an Early Indicator for Risk for Tissue Infection and Wound Chronicity". Annals of Plastic Surgery. 76 (1): 127–31. doi:10.1097/SAP.0000000000000440. PMID 25774966. S2CID 42078581.
  106. ^ Sanclement J, Webster P, Thomas J, Ramadan H (2005). "Bacterial biofilms in surgical specimens of patients with chronic rhinosinusitis". The Laryngoscope. 115 (4): 578–82. doi:10.1097/01.mlg.0000161346.30752.18. PMID 15805862. S2CID 25830188.
  107. ^ Sanderson AR, Leid JG, Hunsaker D (July 2006). "Bacterial biofilms on the sinus mucosa of human subjects with chronic rhinosinusitis". The Laryngoscope. 116 (7): 1121–6. doi:10.1097/01.mlg.0000221954.05467.54. PMID 16826045. S2CID 24785016.
  108. ^ Leevy WM, Gammon ST, Jiang H, Johnson JR, Maxwell DJ, Jackson EN, et al. (December 2006). "Optical imaging of bacterial infection in living mice using a fluorescent near-infrared molecular probe". Journal of the American Chemical Society. 128 (51): 16476–7. doi:10.1021/ja0665592. PMC 2531239. PMID 17177377.
  109. ^ Kaplan JB, Izano EA, Gopal P, Karwacki MT, Kim S, Bose JL, et al. (2012). "Low levels of β-lactam antibiotics induce extracellular DNA release and biofilm formation in Staphylococcus aureus". mBio. 3 (4): e00198-12. doi:10.1128/mBio.00198-12. PMC 3419523. PMID 22851659.
  110. ^ Ibrahim AM (2015). The tragedy of the commons and prisoner's dilemma may improve our realization of the theory of life and provide us with advanced therapeutic ways (Report). doi:10.13140/RG.2.1.2327.9842.
  111. ^ Ciofu O, Tolker-Nielsen T, Jensen PØ, Wang H, Høiby N (May 2015). "Antimicrobial resistance, respiratory tract infections and role of biofilms in lung infections in cystic fibrosis patients". Advanced Drug Delivery Reviews. 85: 7–23. doi:10.1016/j.addr.2014.11.017. PMID 25477303.
  112. ^ Whitchurch CB, Tolker-Nielsen T, Ragas PC, Mattick JS (February 2002). "Extracellular DNA required for bacterial biofilm formation". Science. 295 (5559): 1487. doi:10.1126/science.295.5559.1487. PMID 11859186.
  113. ^ Sen CK, Gordillo GM, Roy S, Kirsner R, Lambert L, Hunt TK, et al. (November 2009). "Human skin wounds: a major and snowballing threat to public health and the economy". Wound Repair and Regeneration. 17 (6): 763–771. doi:10.1111/j.1524-475X.2009.00543.x. PMC 2810192. PMID 19903300.
  114. ^ Singh PK, Parsek MR, Greenberg EP, Welsh MJ (May 2002). "A component of innate immunity prevents bacterial biofilm development". Nature. 417 (6888): 552–555. Bibcode:2002Natur.417..552S. doi:10.1038/417552a. PMID 12037568. S2CID 4423528.
  115. ^ Oggioni MR, Trappetti C, Kadioglu A, Cassone M, Iannelli F, Ricci S, et al. (September 2006). "Switch from planktonic to sessile life: a major event in pneumococcal pathogenesis". Molecular Microbiology. 61 (5): 1196–210. doi:10.1111/j.1365-2958.2006.05310.x. PMC 1618759. PMID 16925554.
  116. ^ Wei H, Håvarstein LS (August 2012). "Fratricide is essential for efficient gene transfer between pneumococci in biofilms". Appl. Environ. Microbiol. 78 (16): 5897–905. Bibcode:2012ApEnM..78.5897W. doi:10.1128/AEM.01343-12. PMC 3406168. PMID 22706053.
  117. ^ Kalsy M, Tonk M, Hardt M, Dobrindt U, Zdybicka-Barabas A, Cytrynska M, Vilcinskas A, Mukherjee K (2020). "The insect antimicrobial peptide cecropin A disrupts uropathogenic Escherichia coli biofilms". npj Biofilms and Microbiomes. 6 (1): 6. doi:10.1038/s41522-020-0116-3. PMC 7016129. PMID 32051417.
  118. ^ Sturbelle RT, de Avila LF, Roos TB, Borchardt JL, da Conceição R, Dellagostin OA, Leite FP (November 2015). "The role of quorum sensing in Escherichia coli (ETEC) virulence factors". Veterinary Microbiology. 180 (3–4): 245–252. doi:10.1016/j.vetmic.2015.08.015. PMID 26386492.
  119. ^ Vogeleer P, Tremblay YD, Mafu AA, Jacques M, Harel J (2014). "Life on the outside: role of biofilms in environmental persistence of Shiga-toxin producing Escherichia coli". Frontiers in Microbiology. 5: 317. doi:10.3389/fmicb.2014.00317. PMC 4076661. PMID 25071733.
  120. ^ Danese PN, Pratt LA, Kolter R (June 2000). "Exopolysaccharide production is required for development of Escherichia coli K-12 biofilm architecture". Journal of Bacteriology. 182 (12): 3593–3596. doi:10.1128/JB.182.12.3593-3596.2000. PMC 101973. PMID 10852895.
  121. ^ Niranjan V, Malini A (June 2014). "Antimicrobial resistance pattern in Escherichia coli causing urinary tract infection among inpatients". The Indian Journal of Medical Research. 139 (6): 945–948. PMC 4165009. PMID 25109731.
  122. ^ Reisner A, Maierl M, Jörger M, Krause R, Berger D, Haid A, et al. (March 2014). "Type 1 fimbriae contribute to catheter-associated urinary tract infections caused by Escherichia coli". Journal of Bacteriology. 196 (5): 931–939. doi:10.1128/JB.00985-13. PMC 3957706. PMID 24336940.
  123. ^ Kobayashi SD, Malachowa N, Whitney AR, Braughton KR, Gardner DJ, Long D, et al. (September 2011). "Comparative analysis of USA300 virulence determinants in a rabbit model of skin and soft tissue infection". The Journal of Infectious Diseases. 204 (6): 937–941. doi:10.1093/infdis/jir441. PMC 3156927. PMID 21849291.
  124. ^ Kitur K, Parker D, Nieto P, Ahn DS, Cohen TS, Chung S, et al. (April 2015). "Toxin-induced necroptosis is a major mechanism of Staphylococcus aureus lung damage". PLOS Pathogens. 11 (4): e1004820. doi:10.1371/journal.ppat.1004820. PMC 4399879. PMID 25880560.
  125. ^ Thurlow LR, Hanke ML, Fritz T, Angle A, Aldrich A, Williams SH, et al. (June 2011). "Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo". Journal of Immunology. 186 (11): 6585–6596. doi:10.4049/jimmunol.1002794. PMC 3110737. PMID 21525381.
  126. ^ Craft KM, Nguyen JM, Berg LJ, Townsend SD (August 2019). "Methicillin-resistant Staphylococcus aureus (MRSA): antibiotic-resistance and the biofilm phenotype". MedChemComm. 10 (8): 1231–1241. doi:10.1039/c9md00044e. PMC 6748282. PMID 31534648.
  127. ^ Lazar V (December 2011). "Quorum sensing in biofilms—how to destroy the bacterial citadels or their cohesion/power?". Anaerobe. 17 (6): 280–5. doi:10.1016/j.anaerobe.2011.03.023. PMID 21497662.
  128. ^ Bjarnsholt T, Jensen PØ, Moser C, Høiby N (2011). Biofilm infections. New York: Springer. ISBN 9781441960832. OCLC 682907381.
  129. ^ a b c d Bryers JD (May 2008). "Medical biofilms". Biotechnology and Bioengineering. 100 (1): 1–18. doi:10.1002/bit.21838. PMC 2706312. PMID 18366134.
  130. ^ Auler ME, Morreira D, Rodrigues FF, Abr Ao MS, Margarido PF, Matsumoto FE, et al. (February 2010). "Biofilm formation on intrauterine devices in patients with recurrent vulvovaginal candidiasis". Medical Mycology. 48 (1): 211–6. doi:10.3109/13693780902856626. PMID 20055746.
  131. ^ Vuotto C, Longo F, Balice MP, Donelli G, Varaldo PE (September 2014). "Antibiotic Resistance Related to Biofilm Formation in Klebsiella pneumoniae". Pathogens. 3 (3): 743–758. doi:10.3390/pathogens3030743. PMC 4243439. PMID 25438022.
  132. ^ Santos-Lopez A, Marshall CW, Scribner MR, Snyder DJ, Cooper VS (September 2019). "Evolutionary pathways to antibiotic resistance are dependent upon environmental structure and bacterial lifestyle". eLife. 8: e47612. doi:10.7554/eLife.47612. PMC 6814407. PMID 31516122.
  133. ^ Pai L, Patil S, Liu S, Wen F (2023). "A growing battlefield in the war against biofilm-induced antimicrobial resistance: insights from reviews on antibiotic resistance". Front Cell Infect Microbiol. 13: 1327069. doi:10.3389/fcimb.2023.1327069. PMC 10770264. PMID 38188636.
  134. ^ a b Kim YW, Subramanian S, Gerasopoulos K, Ben-Yoav H, Wu HC, Quan D, et al. (2015). "Effect of electrical energy on the efficacy of biofilm treatment using the bioelectric effect". npj Biofilms and Microbiomes. 1: 15016. doi:10.1038/npjbiofilms.2015.16. PMC 5515217. PMID 28721233.
  135. ^ Martins dos Santos VA, Yakimov MM, Timmis KN, Golyshin PN (2008). "Genomic Insights into Oil Biodegradation in Marine Systems". In Díaz E (ed.). Microbial Biodegradation: Genomics and Molecular Biology. Horizon Scientific Press. pp. 1971. ISBN 978-1-904455-17-2.
  136. ^ Wang VB, Chua SL, Cai Z, Sivakumar K, Zhang Q, Kjelleberg S, et al. (March 2014). "A stable synergistic microbial consortium for simultaneous azo dye removal and bioelectricity generation". Bioresource Technology. 155: 71–76. doi:10.1016/j.biortech.2013.12.078. PMID 24434696.
  137. ^ Wang VB, Chua SL, Cao B, Seviour T, Nesatyy VJ, Marsili E, et al. (2013). "Engineering PQS biosynthesis pathway for enhancement of bioelectricity production in pseudomonas aeruginosa microbial fuel cells". PLOS ONE. 8 (5): e63129. Bibcode:2013PLoSO...863129W. doi:10.1371/journal.pone.0063129. PMC 3659106. PMID 23700414.
  138. ^ Vera M, Schippers A, Sand W (September 2013). "Progress in bioleaching: fundamentals and mechanisms of bacterial metal sulfide oxidation—part A". Appl. Microbiol. Biotechnol. 97 (17): 7529–41. doi:10.1007/s00253-013-4954-2. PMID 23720034. S2CID 17677624.
  139. ^ Chan SY, Wong MW, Kwan BT, Fang JK, Chua SL (12 October 2022). "Microbial–Enzymatic Combinatorial Approach to Capture and Release Microplastics". Environmental Science & Technology Letters. 9 (11): 975–982. Bibcode:2022EnSTL...9..975C. doi:10.1021/acs.estlett.2c00558. ISSN 2328-8930. S2CID 252892619.
  140. ^ Liu SY, Leung MM, Fang JK, Chua SL (15 January 2021). "Engineering a microbial 'trap and release' mechanism for microplastics removal". Chemical Engineering Journal. 404: 127079. doi:10.1016/j.cej.2020.127079. hdl:10397/88307. ISSN 1385-8947. S2CID 224972583.
  141. ^ a b c d e f g h i Srey S, Jahid ID, Ha SD (June 2013). "Biofilm formation in food industries: A food safety concern". Food Control. 31 (2): 572–585. doi:10.1016/j.foodcont.2012.12.001. ISSN 0956-7135.
  142. ^ T. Tarver, "Biofilms: A Threat to Food Safety – IFT.org", Ift.org, 2016.
  143. ^ a b c d Kumar CG, Anand SK (June 1998). "Significance of microbial biofilms in food industry: a review". International Journal of Food Microbiology. 42 (1–2): 9–27. doi:10.1016/s0168-1605(98)00060-9. PMID 9706794.
  144. ^ Kwok TY, Ma Y, Chua SL (April 2022). "Biofilm dispersal induced by mechanical cutting leads to heightened foodborne pathogen dissemination". Food Microbiology. 102: 103914. doi:10.1016/j.fm.2021.103914. hdl:10397/100037. PMID 34809940. S2CID 244234814.
  145. ^ a b c d Mizan F (2015). "Microbial biofilms in seafood: A food-hygiene challenge". Food Microbiology. 49: 41–55. doi:10.1016/j.fm.2015.01.009. PMID 25846914.
  146. ^ De Araujo LV, Abreu F, Lins U, Santa Anna LM, Nitschke M, Freire DM (January 2011). "Rhamnolipid and surfactin inhibit Listeria monocytogenes adhesion". Food Research International. 44 (1): 481–488. doi:10.1016/j.foodres.2010.09.002.
  147. ^ Wang X, Yao X, Zhu Z, Tang T, Dai K, Sadovskaya I, et al. (July 2009). "Effect of berberine on Staphylococcus epidermidis biofilm formation". International Journal of Antimicrobial Agents. 34 (1): 60–6. doi:10.1016/j.ijantimicag.2008.10.033. PMID 19157797.
  148. ^ Carvalho DB, Fox EG, Santos DG, Sousa JS, Freire DM, Nogueira FC, et al. (July 2019). "Fire Ant Venom Alkaloids Inhibit Biofilm Formation". Toxins. 11 (7): 420. doi:10.3390/toxins11070420. PMC 6669452. PMID 31323790.
  149. ^ Braithwaite RA, McEvoy LA (2004). "Marine biofouling on fish farms and its remediation". Advances in Marine Biology. 47: 215–252. doi:10.1016/S0065-2881(04)47003-5. ISBN 9780120261482. PMID 15596168.
  150. ^ Qian PY, Lau SC, Dahms HU, Dobretsov S, Harder T (2007). "Marine biofilms as mediators of colonization by marine macroorganisms: implications for antifouling and aquaculture". Marine Biotechnology. 9 (4): 399–410. doi:10.1007/s10126-007-9001-9. PMID 17497196. S2CID 7614961.
  151. ^ Cai W, De La Fuente L, Arias CR (September 2013). "Biofilm formation by the fish pathogen Flavobacterium columnare: development and parameters affecting surface attachment". Applied and Environmental Microbiology. 79 (18): 5633–42. Bibcode:2013ApEnM..79.5633C. doi:10.1128/AEM.01192-13. PMC 3754160. PMID 23851087.
  152. ^ King RK, Flick Jr GJ, Pierson D, Smith SA, Boardman GD, Coale Jr CW (2004). "Identification of bacterial pathogens in biofilms of recirculating aquaculture systems". Journal of Aquatic Food Product Technology. 13: 125–133. doi:10.1300/j030v13n01_11. S2CID 83791439.
  153. ^ Bourne DG, Høj L, Webster NS, Swan J, Hall MR (2006). "Biofilm development within a larval rearing tank of the tropical rock lobster, Panulirus ornatus". Aquaculture. 260 (1–4): 27–38. doi:10.1016/j.aquaculture.2006.06.023.
  154. ^ Wietz M, Hall MR, Høj L (July 2009). "Effects of seawater ozonation on biofilm development in aquaculture tanks". Systematic and Applied Microbiology. 32 (4): 266–77. doi:10.1016/j.syapm.2009.04.001. PMID 19446976.
  155. ^ Liu YS, Deng Y, Chen CK, Khoo BL, Chua SL (June 2022). "Rapid detection of microorganisms in a fish infection microfluidics platform". Journal of Hazardous Materials. 431: 128572. doi:10.1016/j.jhazmat.2022.128572. PMID 35278965. S2CID 247136872.
  156. ^ Karunasagar I, Pai R, Malathi G (1994). "Mass mortality of Penaeus monodon larvae due to antibiotic-resistant Vibrio harveyi infection". Aquaculture. 128 (3–4): 203–209. doi:10.1016/0044-8486(94)90309-3.
  157. ^ Lawrence JR, Korber DR, Hoyle BD, Costerton JW, Caldwell DE (October 1991). "Optical sectioning of microbial biofilms". Journal of Bacteriology. 173 (20): 6558–67. doi:10.1128/jb.173.20.6558-6567.1991. PMC 208993. PMID 1917879.
  158. ^ Cooksey K, Wigglesworth-Cooksey B (1995). "Adhesion of bacteria and diatoms to surfaces in the sea: a review". Aquatic Microbial Ecology. 9 (1): 87–96. doi:10.3354/ame009087.
  159. ^ Fanning S, Mitchell AP (2012). "Fungal Biofilms". PLOS Pathog. 8 (4): e1002585. doi:10.1371/journal.ppat.1002585. PMC 3320593. PMID 22496639.
  160. ^ Chandra J, Kuhn DM, Mukherjee PK, Hoyer LL, McCormick T, Ghannoum MA (September 2001). "Biofilm formation by the fungal pathogen Candida albicans: development, architecture, and drug resistance". J. Bacteriol. 183 (18): 5385–94. doi:10.1128/jb.183.18.5385-5394.2001. PMC 95423. PMID 11514524.
  161. ^ Burmølle M, Kjøller A, Sørenses S (2012). Lear G, Gavin L, Lewis G (eds.). Microbial Biofilms: Current Research and Applications. Horizon Scientific Press. pp. 61–71. ISBN 978-1904455967.
  162. ^ Steele DJ, Franklin DJ, Underwood GJ (September 2014). "Protection of cells from salinity stress by extracellular polymeric substances in diatom biofilms". Biofouling. 30 (8): 987–98. doi:10.1080/08927014.2014.960859. PMC 4706044. PMID 25268215.
  163. ^ Windler M, Leinweber K, Bartulos CR, Philipp B, Kroth PG (April 2015). "Biofilm and capsule formation of the diatom Achnanthidium minutissimum are affected by a bacterium". J. Phycol. 51 (2): 343–55. Bibcode:2015JPcgy..51..343W. doi:10.1111/jpy.12280. PMID 26986529. S2CID 1446573.
  164. ^ Buhmann M, Kroth PG, Schleheck D (February 2012). "Photoautotrophic-heterotrophic biofilm communities: a laboratory incubator designed for growing axenic diatoms and bacteria in defined mixed-species biofilms". Environ Microbiol Rep. 4 (1): 133–40. doi:10.1111/j.1758-2229.2011.00315.x. PMID 23757240.
  165. ^ Thomas CM, Nielsen KM (September 2005). "Mechanisms of, and barriers to, horizontal gene transfer between bacteria". Nature Reviews. Microbiology. 3 (9): 711–721. doi:10.1038/nrmicro1234. PMID 16138099. S2CID 1231127.
  166. ^ a b c d e f g h i Luo A, Wang F, Sun D, Liu X, Xin B (2022). "Formation, Development, and Cross-Species Interactions in Biofilms". Frontiers in Microbiology. 12: 757327. doi:10.3389/fmicb.2021.757327. PMC 8764401. PMID 35058893.
  167. ^ Patkowski JB, Dahlberg T, Amin H, Gahlot DK, Vijayrajratnam S, Vogel JP, et al. (April 2023). "The F-pilus biomechanical adaptability accelerates conjugative dissemination of antimicrobial resistance and biofilm formation". Nature Communications. 14 (1): 1879. doi:10.1038/s41467-023-37600-y. PMC 10076315. PMID 37019921.
  168. ^ Thomas VC, Hancock LE (September 2009). "Suicide and fratricide in bacterial biofilms". The International Journal of Artificial Organs. 32 (9): 537–544. doi:10.1177/039139880903200902. PMID 19851979. S2CID 201969291.
  169. ^ a b c d e f g h i j Abe K, Nomura N, Suzuki S (May 2020). "Biofilms: hot spots of horizontal gene transfer (HGT) in aquatic environments, with a focus on a new HGT mechanism". FEMS Microbiology Ecology. 96 (5). doi:10.1093/femsec/fiaa031. PMC 7189800. PMID 32109282.
  170. ^ Chimileski S, Franklin MJ, Papke RT (August 2014). "Biofilms formed by the archaeon Haloferax volcanii exhibit cellular differentiation and social motility, and facilitate horizontal gene transfer". BMC Biology. 12 (1): 65. doi:10.1186/s12915-014-0065-5. PMC 4180959. PMID 25124934.
  171. ^ Azeredo J, Azevedo NF, Briandet R, Cerca N, Coenye T, Costa AR, et al. (May 2017). "Critical review on biofilm methods". Critical Reviews in Microbiology. 43 (3): 313–351. doi:10.1080/1040841X.2016.1208146. hdl:1822/45004. PMID 27868469. S2CID 3991858.

Further reading edit

  • Allison DG (2000). Community structure and co-operation in biofilms. Cambridge, UK: Cambridge University Press. ISBN 978-0-521-79302-5.
  • Lynch JF, Lappin-Scott HM, Costerton JW (2003). Microbial biofilms. Cambridge, UK: Cambridge University Press. ISBN 978-0-521-54212-8.
  • Fratamico M (2009). Biofilms in the food and beverage industries. Woodhead Publishing Limited. ISBN 978-1-84569-477-7.

External links edit

  • A TED-ED animation on basic biofilm biology: The microbial jungles all over the place (and you) by Scott Chimileski and Roberto Kolter
  • Biofilm Archive of Biofilm Research & News 9 October 2010 at the Wayback Machine
  • "Why Am I Still Sick?" – The Movie, 2012: Documentary on Biofilms: The Silent Role of Biofilms in Chronic Disease
  • HD Video Interviews on biofilms, antibiotics, etc. with experts, youtube.com: ADRSupport/biofilm

biofilm, biographic, motion, picture, biographical, film, this, article, relies, excessively, references, primary, sources, please, improve, this, article, adding, secondary, tertiary, sources, find, sources, news, newspapers, books, scholar, jstor, april, 202. For biographic motion picture see Biographical film This article relies excessively on references to primary sources Please improve this article by adding secondary or tertiary sources Find sources Biofilm news newspapers books scholar JSTOR April 2021 Learn how and when to remove this template message A biofilm is an syntrophic community of microorganisms in which cells stick to each other and often also to a surface 2 3 4 These adherent cells become embedded within a slimy extracellular matrix that is composed of extracellular polymeric substances EPSs 3 4 The cells within the biofilm produce the EPS components which are typically a polymeric combination of extracellular polysaccharides proteins lipids and DNA 3 4 5 Because they have three dimensional structure and represent a community lifestyle for microorganisms they have been metaphorically described as cities for microbes 6 7 Staphylococcus aureus biofilm on an indwelling catheterProbable cyanobacteria in the vertical section of a silicified biofilm from the Lower Cretaceous Very shallow hypersaline environment of the Urgonian carbonate platform of Provence south eastern France IUPAC definition Aggregate of microorganisms in which cells that are frequently embedded within a self produced matrix of extracellular polymeric substances EPSs adhere to each other and or to a surface A biofilm is a system that can be adapted internally to environmental conditions by its inhabitants The self produced matrix of extracellular polymeric substances which is also referred to as slime is a polymeric conglomeration generally composed of extracellular biopolymers in various structural forms 1 Biofilms may form on living biotic or non living abiotic surfaces and can be common in natural industrial and hospital settings 4 8 They may constitute a microbiome or be a portion of it The microbial cells growing in a biofilm are physiologically distinct from planktonic cells of the same organism which by contrast are single cells that may float or swim in a liquid medium 9 Biofilms can form on the teeth of most animals as dental plaque where they may cause tooth decay and gum disease Microbes form a biofilm in response to a number of different factors 10 which may include cellular recognition of specific or non specific attachment sites on a surface nutritional cues or in some cases by exposure of planktonic cells to sub inhibitory concentrations of antibiotics 11 12 A cell that switches to the biofilm mode of growth undergoes a phenotypic shift in behavior in which large suites of genes are differentially regulated 13 A biofilm may also be considered a hydrogel which is a complex polymer that contains many times its dry weight in water Biofilms are not just bacterial slime layers but biological systems the bacteria organize themselves into a coordinated functional community Biofilms can attach to a surface such as a tooth or rock and may include a single species or a diverse group of microorganisms Subpopulations of cells within the biofilm differentiate to perform various activities for motility matrix production and sporulation supporting the overall success of the biofilm 14 The biofilm bacteria can share nutrients and are sheltered from harmful factors in the environment such as desiccation antibiotics and a host body s immune system A biofilm usually begins to form when a free swimming planktonic bacterium attaches to a surface 15 page needed Contents 1 Origin and formation 1 1 Origin of biofilms 1 2 Formation of biofilms 2 Development 3 Dispersal 4 Properties 4 1 Extracellular matrix 5 Habitats 5 1 Percolating filters 5 2 Slow sand filter 5 3 Rhizosphere 5 4 Mammalian gut 5 5 Human environment 5 6 Ship fouling 5 7 Stromatolites 5 8 Dental plaque 5 8 1 Intercellular communication 6 Taxonomic diversity 7 Infectious diseases 7 1 Pseudomonas aeruginosa 7 2 Streptococcus pneumoniae 7 3 Escherichia coli 7 4 Staphylococcus aureus 8 Uses and impact 8 1 In medicine 8 2 In industry 8 3 Food industry 8 4 In aquaculture 9 Eukaryotic 10 Horizontal gene transfer 10 1 Examples 11 Cultivation devices 12 See also 13 References 14 Further reading 15 External linksOrigin and formation editOrigin of biofilms edit Biofilms are thought to have arisen during primitive Earth as a defense mechanism for prokaryotes as the conditions at that time were too harsh for their survival They can be found very early in Earth s fossil records about 3 25 billion years ago as both Archaea and Bacteria and commonly protect prokaryotic cells by providing them with homeostasis encouraging the development of complex interactions between the cells in the biofilm 4 Formation of biofilms edit The formation of a biofilm begins with the attachment of free floating microorganisms to a surface 9 6 The first colonist bacteria of a biofilm may adhere to the surface initially by the weak van der Waals forces and hydrophobic effects 16 17 If the colonists are not immediately separated from the surface they can anchor themselves more permanently using cell adhesion structures such as pili A unique group of Archaea that inhabit anoxic groundwater have similar structures called hami Each hamus is a long tube with three hook attachments that are used to attach to each other or to a surface enabling a community to develop 18 19 Hyperthermophilic archaeon Pyrobaculum calidifontis produce bundling pili which are homologous to the bacterial TasA filaments a major component of the extracellular matrix in bacterial biofilms which contribute to biofilm stability 20 TasA homologs are encoded by many other archaea suggesting mechanistic similarities and evolutionary connection between bacterial and archaeal biofilms 20 nbsp Biofilm of golden hydrophobic bacteria ceiling of Golden Dome Cave a lava tube in Lava Beds National Monument 21 Hydrophobicity can also affect the ability of bacteria to form biofilms Bacteria with increased hydrophobicity have reduced repulsion between the substratum and the bacterium 22 Some bacteria species are not able to attach to a surface on their own successfully due to their limited motility but are instead able to anchor themselves to the matrix or directly to other earlier bacteria colonists Non motile bacteria cannot recognize surfaces or aggregate together as easily as motile bacteria 22 During surface colonization bacteria cells are able to communicate using quorum sensing QS products such as N acyl homoserine lactone AHL Once colonization has begun the biofilm grows by a combination of cell division and recruitment Polysaccharide matrices typically enclose bacterial biofilms The matrix exopolysaccharides can trap QS autoinducers within the biofilm to prevent predator detection and ensure bacterial survival 23 In addition to the polysaccharides these matrices may also contain material from the surrounding environment including but not limited to minerals soil particles and blood components such as erythrocytes and fibrin 22 The final stage of biofilm formation is known as development and is the stage in which the biofilm is established and may only change in shape and size citation needed The development of a biofilm may allow for an aggregate cell colony or colonies to be increasingly tolerant 24 or resistant to antibiotics Cell cell communication or quorum sensing has been shown to be involved in the formation of biofilm in several bacterial species 25 Development edit nbsp Mature biofilm structure 26 Biofilm is characterised by heterogenous environment and the presence of a variety of subpopulations A biofilm structure is composed of metabolically active both resistant and tolerant and non active cells viable but not culturable cells and persisters as well as polymer matrix consisting of polysaccharide extracellular DNA and proteins Biofilm growth is associated with an escalated level of mutations and horizontal gene transfer which is promoted in due to the packed and dense structure Bacteria in biofilms communicate by quorum sensing which activates genes participating in virulence factors production 26 27 Biofilms are the product of a microbial developmental process 28 The process is summarized by five major stages of biofilm development as shown in the diagram below 29 nbsp Five stages of biofilm development 29 1 Initial attachment 2 Irreversible attachment 3 Maturation I 4 Maturation II and 5 Dispersion Each stage of development in the diagram is paired with a photomicrograph of a developing P aeruginosa biofilm All photomicrographs are shown to the same scale Dispersal editDispersal of cells from the biofilm colony is an essential stage of the biofilm life cycle Dispersal enables biofilms to spread and colonize new surfaces Enzymes that degrade the biofilm extracellular matrix such as dispersin B and deoxyribonuclease may contribute to biofilm dispersal 30 31 Enzymes that degrade the biofilm matrix may be useful as anti biofilm agents 32 33 Evidence has shown that a fatty acid messenger cis 2 decenoic acid is capable of inducing dispersion and inhibiting growth of biofilm colonies Secreted by Pseudomonas aeruginosa this compound induces cyclo heteromorphic cells in several species of bacteria and the yeast Candida albicans 34 Nitric oxide has also been shown to trigger the dispersal of biofilms of several bacteria species 35 36 at sub toxic concentrations Nitric oxide has potential as a treatment for patients that have chronic infections caused by biofilms 37 It was generally assumed that cells dispersed from biofilms immediately go into the planktonic growth phase However studies have shown that the physiology of dispersed cells from Pseudomonas aeruginosa biofilms is highly different from that of planktonic and biofilm cells 38 39 Hence the dispersal process is a unique stage during the transition from biofilm to planktonic lifestyle in bacteria Dispersed cells are found to be highly virulent against macrophages and Caenorhabditis elegans but highly sensitive towards iron stress as compared with planktonic cells 38 Furthermore Pseudomonas aeruginosa biofilms undergo distinct spatiotemporal dynamics during biofilm dispersal or disassembly with contrasting consequences in recolonization and disease dissemination 40 Biofilm dispersal induced bacteria to activate dispersal genes to actively depart from biofilms as single cells at consistent velocities but could not recolonize fresh surfaces In contrast biofilm disassembly by degradation of a biofilm exopolysaccharide released immotile aggregates at high initial velocities enabling the bacteria to recolonize fresh surfaces and cause infections in the hosts efficiently Hence biofilm dispersal is more complex than previously thought where bacterial populations adopting distinct behavior after biofilm departure may be the key to survival of bacterial species and dissemination of diseases nbsp Biofilm dispersal Properties editBiofilms are usually found on solid substrates submerged in or exposed to an aqueous solution although they can form as floating mats on liquid surfaces and also on the surface of leaves particularly in high humidity climates Given sufficient resources for growth a biofilm will quickly grow to be macroscopic visible to the naked eye Biofilms can contain many different types of microorganism e g bacteria archaea protozoa fungi and algae each group performs specialized metabolic functions However some organisms will form single species films under certain conditions The social structure cooperation competition within a biofilm depends highly on the different species present 41 Extracellular matrix edit nbsp Scanning electron micrograph of mixed culture biofilm demonstrating in detail a spatially heterogeneous arrangement of bacterial cells and extracellular polymeric substances The EPS matrix consists of exopolysaccharides proteins and nucleic acids 42 43 44 A large proportion of the EPS is more or less strongly hydrated however hydrophobic EPS also occur one example is cellulose 45 which is produced by a range of microorganisms This matrix encases the cells within it and facilitates communication among them through biochemical signals as well as gene exchange The EPS matrix also traps extracellular enzymes and keeps them in close proximity to the cells Thus the matrix represents an external digestion system and allows for stable synergistic microconsortia of different species 46 Some biofilms have been found to contain water channels that help distribute nutrients and signalling molecules 47 This matrix is strong enough that under certain conditions biofilms can become fossilized stromatolites Bacteria living in a biofilm usually have significantly different properties from free floating bacteria of the same species as the dense and protected environment of the film allows them to cooperate and interact in various ways 48 One benefit of this environment is increased resistance to detergents and antibiotics as the dense extracellular matrix and the outer layer of cells protect the interior of the community 49 50 In some cases antibiotic resistance can be increased up to 5 000 times 51 Lateral gene transfer is often facilitated within bacterial and archaeal biofilms 52 and can leads to a more stable biofilm structure 53 Extracellular DNA is a major structural component of many different microbial biofilms 54 Enzymatic degradation of extracellular DNA can weaken the biofilm structure and release microbial cells from the surface However biofilms are not always less susceptible to antibiotics For instance the biofilm form of Pseudomonas aeruginosa has no greater resistance to antimicrobials than do stationary phase planktonic cells although when the biofilm is compared to logarithmic phase planktonic cells the biofilm does have greater resistance to antimicrobials This resistance to antibiotics in both stationary phase cells and biofilms may be due to the presence of persister cells 55 Habitats edit nbsp Mats of bacterial biofilm color the hot springs in Yellowstone National Park The longest raised mat area is about half a meter long nbsp Thermophilic bacteria in the outflow of Mickey Hot Springs Oregon approximately 20 mm thick Biofilms are ubiquitous in organic life Nearly every species of microorganism have mechanisms by which they can adhere to surfaces and to each other Biofilms will form on virtually every non shedding surface in non sterile aqueous or humid environments Biofilms can grow in the most extreme environments from for example the extremely hot briny waters of hot springs ranging from very acidic to very alkaline to frozen glaciers Biofilms can be found on rocks and pebbles at the bottoms of most streams or rivers and often form on the surfaces of stagnant pools of water Biofilms are important components of food chains in rivers and streams and are grazed by the aquatic invertebrates upon which many fish feed Biofilms are found on the surface of and inside plants They can either contribute to crop disease or as in the case of nitrogen fixing rhizobia on root nodules exist symbiotically with the plant 56 Examples of crop diseases related to biofilms include citrus canker Pierce s disease of grapes and bacterial spot of plants such as peppers and tomatoes 57 Percolating filters edit Percolating filters in sewage treatment works are highly effective removers of pollutants from settled sewage liquor They work by trickling the liquid over a bed of hard material which is designed to have a very large surface area A complex biofilm develops on the surface of the medium which absorbs adsorbs and metabolises the pollutants The biofilm grows rapidly and when it becomes too thick to retain its grip on the media it washes off and is replaced by newly grown film The washed off sloughed off film is settled out of the liquid stream to leave a highly purified effluent 58 Slow sand filter edit Slow sand filters are used in water purification for treating raw water to produce a potable product They work through the formation of a biofilm called the hypogeal layer or Schmutzdecke in the top few millimetres of the fine sand layer The Schmutzdecke is formed in the first 10 20 days of operation 59 and consists of bacteria fungi protozoa rotifera and a range of aquatic insect larvae As an epigeal biofilm ages more algae tend to develop and larger aquatic organisms may be present including some bryozoa snails and annelid worms The surface biofilm is the layer that provides the effective purification in potable water treatment the underlying sand providing the support medium for this biological treatment layer As water passes through the hypogeal layer particles of foreign matter are trapped in the mucilaginous matrix and soluble organic material is adsorbed The contaminants are metabolised by the bacteria fungi and protozoa The water produced from an exemplary slow sand filter is of excellent quality with 90 99 bacterial cell count reduction 60 Rhizosphere edit Plant beneficial microbes can be categorized as plant growth promoting rhizobacteria 61 These plant growth promoters colonize the roots of plants and provide a wide range of beneficial functions for their host including nitrogen fixation pathogen suppression anti fungal properties and the breakdown of organic materials 62 One of these functions is the defense against pathogenic soil borne bacteria and fungi by way of induced systemic resistance ISR 63 or induced systemic responses triggered by pathogenic microbes pathogen induced systemic acquired resistance 64 Plant exudates act as chemical signals for host specific bacteria to colonize 65 Rhizobacteria colonization steps include attractions recognition adherence colonization and growth 62 Bacteria that have been shown to be beneficial and form biofilms include Bacillus Pseudomonas and Azospirillum 66 67 Biofilms in the rhizosphere often result in pathogen or plant induced systemic resistances Molecular properties on the surface of the bacterium cause an immune response in the plant host 65 These microbe associated molecules interact with receptors on the surface of plant cells and activate a biochemical response that is thought to include several different genes at a number of loci 65 Several other signaling molecules have been linked to both induced systemic responses and pathogen induced systemic responses such as jasmonic acid and ethylene 62 Cell envelope components such as bacterial flagella and lipopolysaccharides which are recognized by plant cells as components of pathogens 68 Certain iron metabolites produced by Pseudomonas have also been shown to create an induced systemic response 65 This function of the biofilm helps plants build stronger resistance to pathogens Plants that have been colonized by PGPR forming a biofilm have gained systemic resistances and are primed for defense against pathogens This means that the genes necessary for the production of proteins that work towards defending the plant against pathogens have been expressed and the plant has a stockpile of compounds to release to fight off pathogens 65 A primed defense system is much faster in responding to pathogen induced infection and may be able to deflect pathogens before they are able to establish themselves 69 Plants increase the production of lignin reinforcing cell walls and making it difficult for pathogens to penetrate into the cell while also cutting off nutrients to already infected cells effectively halting the invasion 62 They produce antimicrobial compounds such as phytoalexins chitinases and proteinase inhibitors which prevent the growth of pathogens 64 These functions of disease suppression and pathogen resistance ultimately lead to an increase in agricultural production and a decrease in the use of chemical pesticides herbicides and fungicides because there is a reduced amount of crop loss due to disease 70 Induced systemic resistance and pathogen induced systemic acquired resistance are both potential functions of biofilms in the rhizosphere and should be taken into consideration when applied to new age agricultural practices because of their effect on disease suppression without the use of dangerous chemicals Mammalian gut edit Studies in 2003 discovered that the immune system supports biofilm development in the large intestine This was supported mainly with the fact that the two most abundantly produced molecules by the immune system also support biofilm production and are associated with the biofilms developed in the gut This is especially important because the appendix holds a mass amount of these bacterial biofilms 71 This discovery helps to distinguish the possible function of the appendix and the idea that the appendix can help reinoculate the gut with good gut flora However modified or disrupted states of biofilms in the gut have been connected to diseases such as inflammatory bowel disease and colorectal cancer 72 Human environment edit In the human environment biofilms can grow in showers very easily since they provide a moist and warm environment for them to thrive They can form inside water and sewage pipes and cause clogging and corrosion On floors and counters they can make sanitation difficult in food preparation areas In soil they can cause bioclogging In cooling or heating water systems they are known to reduce heat transfer 73 Biofilms in marine engineering systems such as pipelines of the offshore oil and gas industry 74 can lead to substantial corrosion problems Corrosion is mainly due to abiotic factors however at least 20 of corrosion is caused by microorganisms that are attached to the metal subsurface i e microbially influenced corrosion Ship fouling edit Bacterial adhesion to boat hulls serves as the foundation for biofouling of seagoing vessels Once a film of bacteria forms it is easier for other marine organisms such as barnacles to attach Such fouling can reduce maximum vessel speed by up to 20 prolonging voyages and consuming fuel Time in dry dock for refitting and repainting reduces the productivity of shipping assets and the useful life of ships is also reduced due to corrosion and mechanical removal scraping of marine organisms from ships hulls Stromatolites edit Stromatolites are layered accretionary structures formed in shallow water by the trapping binding and cementation of sedimentary grains by microbial biofilms especially of cyanobacteria Stromatolites include some of the most ancient records of life on Earth and are still forming today Dental plaque edit Within the human body biofilms are present on the teeth as dental plaque where they may cause tooth decay and gum disease These biofilms can either be in an uncalcified state that can be removed by dental instruments or a calcified state which is more difficult to remove Removal techniques can also include antimicrobials 75 Dental plaque is an oral biofilm that adheres to the teeth and consists of many species of both bacteria and fungi such as Streptococcus mutans and Candida albicans embedded in salivary polymers and microbial extracellular products The accumulation of microorganisms subjects the teeth and gingival tissues to high concentrations of bacterial metabolites which results in dental disease 76 Biofilm on the surface of teeth is frequently subject to oxidative stress 77 and acid stress 78 Dietary carbohydrates can cause a dramatic decrease in pH in oral biofilms to values of 4 and below acid stress 78 A pH of 4 at body temperature of 37 C causes depurination of DNA leaving apurinic AP sites in DNA 79 especially loss of guanine 80 Dental plaque biofilm can result in dental caries if it is allowed to develop over time An ecologic shift away from balanced populations within the dental biofilm is driven by certain cariogenic microbiological populations beginning to dominate when the environment favors them The shift to an acidogenic aciduric and cariogenic microbiological population develops and is maintained by frequent consumption of fermentable dietary carbohydrate The resulting activity shift in the biofilm and resulting acid production within the biofilm at the tooth surface is associated with an imbalance of demineralization over remineralization leading to net mineral loss within dental hard tissues enamel and then dentin the symptom being a carious lesion or cavity By preventing the dental plaque biofilm from maturing or by returning it back to a non cariogenic state dental caries can be prevented and arrested 81 82 This can be achieved through the behavioral step of reducing the supply of fermentable carbohydrates i e sugar intake and frequent removal of the biofilm i e toothbrushing 81 Intercellular communication edit See also Oral microbiology A peptide pheromone quorum sensing signaling system in S mutans includes the competence stimulating peptide CSP that controls genetic competence 83 84 Genetic competence is the ability of a cell to take up DNA released by another cell Competence can lead to genetic transformation a form of sexual interaction favored under conditions of high cell density and or stress where there is maximal opportunity for interaction between the competent cell and the DNA released from nearby donor cells This system is optimally expressed when S mutans cells reside in an actively growing biofilm Biofilm grown S mutans cells are genetically transformed at a rate 10 to 600 fold higher than S mutans growing as free floating planktonic cells suspended in liquid 83 When the biofilm containing S mutans and related oral streptococci is subjected to acid stress the competence regulon is induced leading to resistance to being killed by acid 78 As pointed out by Michod et al transformation in bacterial pathogens likely provides for effective and efficient recombinational repair of DNA damages 85 It appears that S mutans can survive the frequent acid stress in oral biofilms in part through the recombinational repair provided by competence and transformation Predator prey interactions See also Predator prey interactions Predator prey interactions between biofilms and bacterivores such as the soil dwelling nematode Caenorhabditis elegans had been extensively studied Via the production of sticky matrix and formation of aggregates Yersinia pestis biofilms can prevent feeding by obstructing the mouth of C elegans 86 Moreover Pseudomonas aeruginosa biofilms can impede the slithering motility of C elegans termed as quagmire phenotype resulting in trapping of C elegans within the biofilms and preventing the exploration of nematodes to feed on susceptible biofilms 87 This significantly reduced the ability of predator to feed and reproduce thereby promoting the survival of biofilms Pseudomonas aeruginosa biofilms can also mask their chemical signatures where they reduced the diffusion of quorum sensing molecules into the environment and prevented the detection of C elegans 88 Taxonomic diversity editMany different bacteria form biofilms including gram positive e g Bacillus spp Listeria monocytogenes Staphylococcus spp and lactic acid bacteria including Lactobacillus plantarum and Lactococcus lactis and gram negative species e g Escherichia coli or Pseudomonas aeruginosa 89 Cyanobacteria also form biofilms in aquatic environments 90 Biofilms are formed by bacteria that colonize plants e g Pseudomonas putida Pseudomonas fluorescens and related pseudomonads which are common plant associated bacteria found on leaves roots and in the soil and the majority of their natural isolates form biofilms 91 Several nitrogen fixing symbionts of legumes such as Rhizobium leguminosarum and Sinorhizobium meliloti form biofilms on legume roots and other inert surfaces 91 Along with bacteria biofilms are also generated by archaea 52 and by a range of eukaryotic organisms including fungi e g Cryptococcus laurentii 92 and microalgae Among microalgae one of the main progenitors of biofilms are diatoms which colonise both fresh and marine environments worldwide 93 94 For other species in disease associated biofilms and biofilms arising from eukaryotes see below Infectious diseases editBiofilms have been found to be involved in a wide variety of microbial infections in the body by one estimate 80 of all infections 95 Infectious processes in which biofilms have been implicated include common problems such as bacterial vaginosis urinary tract infections catheter infections middle ear infections formation of dental plaque 96 gingivitis coating contact lenses 97 and less common but more lethal processes such as endocarditis infections in cystic fibrosis and infections of permanent indwelling devices such as joint prostheses heart valves and intervertebral disc 98 99 100 The first visual evidence of a biofilm was recorded after spine surgery 101 It was found that in the absence of clinical presentation of infection impregnated bacteria could form a biofilm around an implant and this biofilm can remain undetected via contemporary diagnostic methods including swabbing Implant biofilm is frequently present in aseptic pseudarthrosis cases 101 102 103 Furthermore it has been noted that bacterial biofilms may impair cutaneous wound healing and reduce topical antibacterial efficiency in healing or treating infected skin wounds 104 The diversity of P aeruginosa cells within a biofilm is thought to make it harder to treat the infected lungs of people with cystic fibrosis 14 Early detection of biofilms in wounds is crucial to successful chronic wound management Although many techniques have developed to identify planktonic bacteria in viable wounds few have been able to quickly and accurately identify bacterial biofilms Future studies are needed to find means of identifying and monitoring biofilm colonization at the bedside to permit timely initiation of treatment 105 It has been shown that biofilms are present on the removed tissue of 80 of patients undergoing surgery for chronic sinusitis The patients with biofilms were shown to have been denuded of cilia and goblet cells unlike the controls without biofilms who had normal cilia and goblet cell morphology 106 Biofilms were also found on samples from two of 10 healthy controls mentioned The species of bacteria from intraoperative cultures did not correspond to the bacteria species in the biofilm on the respective patient s tissue In other words the cultures were negative though the bacteria were present 107 New staining techniques are being developed to differentiate bacterial cells growing in living animals e g from tissues with allergy inflammations 108 Research has shown that sub therapeutic levels of b lactam antibiotics induce biofilm formation in Staphylococcus aureus This sub therapeutic level of antibiotic may result from the use of antibiotics as growth promoters in agriculture or during the normal course of antibiotic therapy The biofilm formation induced by low level methicillin was inhibited by DNase suggesting that the sub therapeutic levels of antibiotic also induce extracellular DNA release 109 Moreover from an evolutionary point of view the creation of the tragedy of the commons in pathogenic microbes may provide advanced therapeutic ways for chronic infections caused by biofilms via genetically engineered invasive cheaters who can invade wild types cooperators of pathogenic bacteria until cooperator populations go to extinction or overall population cooperators and cheaters go to extinction 110 Pseudomonas aeruginosa edit P aeruginosa represents a commonly used biofilm model organism since it is involved in different types of biofilm associated chronic infections 42 Examples of such infections include chronic wounds chronic otitis media chronic prostatitis and chronic lung infections in cystic fibrosis CF patients About 80 of CF patients have chronic lung infection caused mainly by P aeruginosa growing in a non surface attached biofilms surround by PMN 111 The infection remains present despite aggressive antibiotic therapy and is a common cause of death in CF patients due to constant inflammatory damage to the lungs 42 In patients with CF one therapy for treating early biofilm development is to employ DNase to structurally weaken the biofilm 5 112 Biofilm formation of P aeruginosa along with other bacteria is found in 90 of chronic wound infections which leads to poor healing and high cost of treatment estimated at more than US 25 billion every year in the United States 113 In order to minimize the P aeruginosa infection host epithelial cells secrete antimicrobial peptides such as lactoferrin to prevent the formation of the biofilms 114 Streptococcus pneumoniae edit Streptococcus pneumoniae is the main cause of community acquired pneumonia and meningitis in children and the elderly and of sepsis in HIV infected persons When S pneumoniae grows in biofilms genes are specifically expressed that respond to oxidative stress and induce competence 115 Formation of a biofilm depends on competence stimulating peptide CSP CSP also functions as a quorum sensing peptide It not only induces biofilm formation but also increases virulence in pneumonia and meningitis It has been proposed that competence development and biofilm formation is an adaptation of S pneumoniae to survive the defenses of the host 85 In particular the host s polymorphonuclear leukocytes produce an oxidative burst to defend against the invading bacteria and this response can kill bacteria by damaging their DNA Competent S pneumoniae in a biofilm have the survival advantage that they can more easily take up transforming DNA from nearby cells in the biofilm to use for recombinational repair of oxidative damages in their DNA Competent S pneumoniae can also secrete an enzyme murein hydrolase that destroys non competent cells fratricide causing DNA to be released into the surrounding medium for potential use by the competent cells 116 The insect antimicrobial peptide cecropin A can destroy planktonic and sessile biofilm forming uropathogenic E coli cells either alone or when combined with the antibiotic nalidixic acid synergistically clearing infection in vivo in the insect host Galleria mellonella without off target cytotoxicity The multi target mechanism of action involves outer membrane permeabilization followed by biofilm disruption triggered by the inhibition of efflux pump activity and interactions with extracellular and intracellular nucleic acids 117 Escherichia coli edit Escherichia coli biofilms are responsible for many intestinal infectious diseases 118 The Extraintestinal group of E coli ExPEC is the dominant bacterial group that attacks the urinary system which leads to urinary tract infections 119 The biofilm formation of these pathogenic E coli is hard to eradicate due to the complexity of its aggregation structure and it has a significant contribution to developing aggressive medical complications increase in hospitalization rate and cost of treatment 120 121 The development of E coli biofilm is a common leading cause of urinary tract infections UTI in hospitals through its contribution to developing medical device associated infections Catheter associated urinary tract infections CAUTI represent the most common hospital acquired infection due to the formation of the pathogenic E coli biofilm inside the catheters 122 Staphylococcus aureus edit Staphylococcus aureus pathogen can attack skin and lungs leading to skin infection and pneumonia 123 124 Moreover the biofilm infections network of S aureus plays a critical role in preventing immune cells such as macrophages from eliminating and destroying bacterial cells 125 Furthermore biofilm formation by bacteria such as S aureus not only develops resistance against antibiotic medication but also develop internal resistance toward antimicrobial peptides AMPs leading to preventing the inhibition of the pathogen and maintaining its survival 126 Uses and impact editIn medicine edit It is suggested that around two thirds of bacterial infections in humans involve biofilms 51 127 Infections associated with the biofilm growth usually are challenging to eradicate 128 This is mostly due to the fact that mature biofilms display antimicrobial tolerance and immune response evasions 129 42 Biofilms often form on the inert surfaces of implanted devices such as catheters prosthetic cardiac valves and intrauterine devices 130 Some of the most difficult infections to treat are those associated with the use of medical devices 51 102 The rapidly expanding worldwide industry for biomedical devices and tissue engineering related products is already at 180 billion per year yet this industry continues to suffer from microbial colonization No matter the sophistication microbial infections can develop on all medical devices and tissue engineering constructs 129 60 70 of hospital acquired infections are associated with the implantation of a biomedical device 129 This leads to 2 million cases annually in the U S costing the healthcare system over 5 billion in additional healthcare expenses 129 The level of antibiotic resistance in a biofilm is much greater than that of non biofilm bacteria and can be as much as 5 000 times greater 51 The extracellular matrix of biofilm is considered one of the leading factors that can reduce the penetration of antibiotics into a biofilm structure and contributes to antibiotic resistance 131 Further it has been demonstrated that the evolution of resistance to antibiotics may be affected by the biofilm lifestyle 132 Bacteriophage therapy can disperse the biofilm generated by antibiotic resistant bacteria 133 It has been shown that the introduction of a small current of electricity to the liquid surrounding a biofilm together with small amounts of antibiotic can reduce the level of antibiotic resistance to levels of non biofilm bacteria This is termed the bioelectric effect 51 134 The application of a small DC current on its own can cause a biofilm to detach from its surface 51 A study showed that the type of current used made no difference to the bioelectric effect 134 In industry edit See also Biofilms in industry Biofilms can also be harnessed for constructive purposes For example many sewage treatment plants include a secondary treatment stage in which waste water passes over biofilms grown on filters which extract and digest organic compounds In such biofilms bacteria are mainly responsible for removal of organic matter BOD while protozoa and rotifers are mainly responsible for removal of suspended solids SS including pathogens and other microorganisms Slow sand filters rely on biofilm development in the same way to filter surface water from lake spring or river sources for drinking purposes What is regarded as clean water is effectively a waste material to these microcellular organisms Biofilms can help eliminate petroleum oil from contaminated oceans or marine systems The oil is eliminated by the hydrocarbon degrading activities of communities of hydrocarbonoclastic bacteria HCB 135 Biofilms are used in microbial fuel cells MFCs to generate electricity from a variety of starting materials including complex organic waste and renewable biomass 8 136 137 Biofilms are also relevant for the improvement of metal dissolution in bioleaching industry 138 and aggregation of microplastics pollutants for convenient removal from the environment 139 140 Food industry edit Biofilms have become problematic in several food industries due to the ability to form on plants and during industrial processes 141 Bacteria can survive long periods of time in water animal manure and soil causing biofilm formation on plants or in the processing equipment 142 The buildup of biofilms can affect the heat flow across a surface and increase surface corrosion and frictional resistance of fluids 143 These can lead to a loss of energy in a system and overall loss of products 143 Along with economic problems biofilm formation on food poses a health risk to consumers due to the ability to make the food more resistant to disinfectants 141 As a result from 1996 to 2010 the Centers for Disease Control and Prevention estimated 48 million foodborne illnesses per year 141 Biofilms have been connected to about 80 of bacterial infections in the United States 141 In produce microorganisms attach to the surfaces and biofilms develop internally 141 During the washing process biofilms resist sanitization and allow bacteria to spread across the produce 141 especially via kitchen utensils 144 This problem is also found in ready to eat foods because the foods go through limited cleaning procedures before consumption 141 Due to the perishability of dairy products and limitations in cleaning procedures resulting in the buildup of bacteria dairy is susceptible to biofilm formation and contamination 141 143 The bacteria can spoil the products more readily and contaminated products pose a health risk to consumers One species of bacteria that can be found in various industries and is a major cause of foodborne disease is Salmonella 145 Large amounts of Salmonella contamination can be found in the poultry processing industry as about 50 of Salmonella strains can produce biofilms on poultry farms 141 Salmonella increases the risk of foodborne illnesses when the poultry products are not cleaned and cooked correctly Salmonella is also found in the seafood industry where biofilms form from seafood borne pathogens on the seafood itself as well as in water 145 Shrimp products are commonly affected by Salmonella because of unhygienic processing and handling techniques 145 The preparation practices of shrimp and other seafood products can allow for bacteria buildup on the products 145 New forms of cleaning procedures are being tested to reduce biofilm formation in these processes which will lead to safer and more productive food processing industries These new forms of cleaning procedures also have a profound effect on the environment often releasing toxic gases into the groundwater reservoirs 143 As a response to the aggressive methods employed in controlling biofilm formation there are a number of novel technologies and chemicals under investigation that can prevent either the proliferation or adhesion of biofilm secreting microbes Latest proposed biomolecules presenting marked anti biofilm activity include a range of metabolites such as bacterial rhamnolipids 146 and even plant 147 and animal derived alkaloids 148 In aquaculture edit Further information Aquaculture nbsp A biofilm from the Dead SeaIn shellfish and algal aquaculture biofouling microbial species tend to block nets and cages and ultimately outcompete the farmed species for space and food 149 Bacterial biofilms start the colonization process by creating microenvironments that are more favorable for biofouling species In the marine environment biofilms could reduce the hydrodynamic efficiency of ships and propellers lead to pipeline blockage and sensor malfunction and increase the weight of appliances deployed in seawater 150 Numerous studies have shown that biofilm can be a reservoir for potentially pathogenic bacteria in freshwater aquaculture 151 152 153 154 Moreover biofilms are important in establishing infections on the fish 155 As mentioned previously biofilms can be difficult to eliminate even when antibiotics or chemicals are used in high doses 156 157 The role that biofilm plays as reservoirs of bacterial fish pathogens has not been explored in detail but it certainly deserves to be studied Eukaryotic editSee also Phototrophic biofilms Along with bacteria biofilms are often initiated and produced by eukaryotic microbes The biofilms produced by eukaryotes is usually occupied by bacteria and other eukaryotes alike however the surface is cultivated and EPS is secreted initially by the eukaryote 92 93 158 Both fungi and microalgae are known to form biofilms in such a way Biofilms of fungal origin are important aspects of human infection and fungal pathogenicity as the fungal infection is more resistant to antifungals 159 160 In the environment fungal biofilms are an area of ongoing research One key area of research is fungal biofilms on plants For example in the soil plant associated fungi including mycorrhiza have been shown to decompose organic matter and protect plants from bacterial pathogens 161 Biofilms in aquatic environments are often founded by diatoms The exact purpose of these biofilms is unknown however there is evidence that the EPS produced by diatoms facilitates both cold and salinity stress 94 162 These eukaryotes interact with a diverse range of other organisms within a region known as the phycosphere but importantly are the bacteria associated with diatoms as it has been shown that although diatoms excrete EPS they only do so when interacting with certain bacteria species 163 164 Horizontal gene transfer editHorizontal gene transfer is the lateral transfer of genetic material between cellular organisms It happens frequently in prokaryotes and less frequently in eukaryotes In bacteria horizontal gene transfer can occur through transformation uptake of free floating DNA in the environment transduction virus mediated DNA uptake or conjugation transfer of DNA between pili structures of two adjacent bacteria 165 Recent studies have also uncovered other mechanisms such as membrane vesicle transmission or gene transfer agents 166 Biofilms promote horizontal gene transfer in a variety of ways nbsp Transmission electron micrograph showing bacteria Escherichia coli that form extensive biofilms using a network of conjugative F pili Source Jonasz PatkowskiBacterial conjugation has been shown to accelerate biofilm formation in difficult environment due to the robust connections established by the conjugative pili 167 These connections can often foster cross species transfer events due to the diverse heterogeneity of many biofilms Additionally biofilms are structurally confined by a polysaccharide matrix providing the close spatial requirements for conjugation Transformation is also frequently observed in biofilms Bacterial autolysis is a key mechanism in biofilm structural regulation providing an abundant source of competent DNA primed for transformative uptake 168 166 In some instances inter biofilm quorum sensing can enhance the competence of free floating eDNA further promoting transformation 166 Stx gene transfer through bacteriophage carriers has been witnessed within biofilms which suggests that biofilms are also a suitable environment for transduction 166 Membrane vesicles HGT occurs when released membrane vesicles containing genetic information fuse with a recipient bacteria and release genetic material into the bacteria s cytoplasm 166 Recent research has revealed that membrane vesicle HGT can promote single strain biofilm formation yet the role membrane vesicle HGT plays in the formation of multistrain biofilms is still unknown 166 GTAs or gene transfer agents are phage like particles produced by the host bacteria and contain random DNA fragments from the host bacteria genome 166 HGT within biofilms can confer antibiotic resistance or increased pathogenicity across the biofilms population promoting biofilm homeostasis 166 Examples edit Conjugative plasmids may encode biofilm associated proteins such as PtgA PrgB or PrgC which promote cell adhesion required for early biofilm formation 169 Genes encoding type III fimbriae are found in pOLA52 Klebsiella pneumoniae plasmid which promote conjugative pilus dependent biofilm formation 169 Transformation commonly occurs within biofilms A phenomenon called fratricide can be seen among streptococcal species in which cell wall degrading enzymes are released lysing neighboring bacteria and releasing their DNA This DNA can then be taken up by the surviving bacteria transformation 169 Competence stimulating peptides may play an important role in biofilm formation among S pneumoniae and S mutans as well 169 Among V cholerae the competence pilus itself promotes cell aggregation through pilus pilus interactions at the beginning of biofilm formation 169 Phage invasion may play a role in biofilm life cycles lysing bacteria and releasing their eDNA which strengthens biofilm structures and can be taken up by neighboring bacteria in transformation 169 Biofilm destruction caused by the E coli phage Rac and the P aeruginosa prophage Pf4 causes detachment of cells from the biofilm 169 Detachment is a biofilm phenomenon which requires more study but is hypothesized to proliferate the bacterial species that comprise the biofilm Membrane vesicle HGT has been witnessed occurring in marine environments among Neisseria gonorrhoeae Pseudomonas aeruginosa Helicobacter pylori and among many other bacterial species 169 Even though membrane vesicle HGT has been shown as a contributing factor in biofilm formation research is still required to prove that membrane vesicle mediated HGT occurs within biofilms 166 169 Membrane vesicle HGT has also been shown to modulate phage bacteria interactions in Bacillus subtilis SPP1 phage resistant cells lacking the SPP1 receptor protein Upon exposure to vesicles containing receptors transduction of pBT163 a cat encoding plasmid occurs resulting in the expression of the SPP1 receptor protein opening the receptive bacteria to future phage infection 169 Recent research has shown that the archaeal species H volcanii has some biofilm phenotypes similar to bacterial biofilms such as differentiation and HGT which required cell cell contact and involved formation of cytosolic bridges and cellular fusion events 170 Cultivation devices editThere is a wide variety of biofilm cultivation devices to mimic natural or industrial environments Although it is important to consider that the particular experimental platform for biofilm research determines what kind of biofilm is cultivated and the data that can be extracted These devices can be grouped into the following 171 microtiter plate MTP systems and MBEC Assay formerly the Calgary Biofilm Device CBD BioFilm Ring Test Archived 26 October 2021 at the Wayback Machine BRT or clinical Biofilm Ring Test cBRT Robbins Device or modified Robbins Device such as the MPMR 10PMMA or the Bio inLine Biofilm Reactor Drip Flow Biofilm Reactor rotary devices such as the CDC Biofilm Reactor the Rotating Disk Reactor the Biofilm Annular Reactor the Industrial Surfaces Biofilm Reactor or the Constant Depth Film Fermenter flow chambers or flow cells such as the Coupon Evaluation Flow Cell Transmission Flow Cell and Capillary Flow Cell from BioSurface Technologies microfluidic approaches such as 3D bacterial biofilm dispersal then recolonization BDR microfluidic model 40 See also edit nbsp Biology portalAmyloid Insoluble protein aggregate with a fibrillar morphology Bacterial nanowires Electrically conductive appendages produced by a number of bacteria Biofilm factory The use of microbial biofilms for chemical productionPages displaying short descriptions of redirect targets Biofilm prevention Stopping microbes attaching to a surface Biomineralization Process by which living organisms produce minerals Center for Biofilm Engineering interdisciplinary research education and technology transfer institute of Montana State UniversityPages displaying wikidata descriptions as a fallback Curli A proteinaceous extracellular fiber produced by enteric bacteria Floc biofilm Type of microbial aggregate suspension Microbial intelligence Adaptive behavior by microscopic organisms including bacteria and protists Microbial mat Multi layered sheet of microorganisms Phage therapy Therapeutic use of bacteriophages to treat bacterial infections Phototrophic biofilm Microbial communities including microorganisms which use light as their energy source Stromatolite Layered sedimentary structure formed by the growth of bacteria or algaeReferences edit Vert M Doi Y Hellwich KH Hess M Hodge P Kubisa P Rinaudo M Schue F 2012 Terminology for biorelated polymers and applications IUPAC Recommendations 2012 Pure and Applied Chemistry 84 2 377 410 doi 10 1351 PAC REC 10 12 04 Biofilms an overview ScienceDirect Topics www sciencedirect com Retrieved 28 January 2024 a b c Lopez D Vlamakis H Kolter R July 2010 Biofilms Cold Spring Harbor Perspectives in Biology 2 7 a000398 doi 10 1101 cshperspect a000398 PMC 2890205 PMID 20519345 a b c d e Hall Stoodley L Costerton JW Stoodley P February 2004 Bacterial biofilms from the natural environment to infectious diseases Nature Reviews Microbiology 2 2 95 108 doi 10 1038 nrmicro821 PMID 15040259 S2CID 9107205 a b Aggarwal S Stewart PS Hozalski RM January 2016 Biofilm Cohesive Strength as a Basis for Biofilm Recalcitrance Are Bacterial Biofilms Overdesigned Microbiology Insights 8 Suppl 2 29 32 doi 10 4137 MBI S31444 PMC 4718087 PMID 26819559 a b Watnick P Kolter R May 2000 Biofilm city of microbes Journal of Bacteriology 182 10 2675 9 doi 10 1128 jb 182 10 2675 2679 2000 PMC 101960 PMID 10781532 Building Codes for Bacterial Cities Quanta Magazine Quanta Magazine Retrieved 25 July 2017 a b Lear G Lewis GD eds 2012 Microbial Biofilms Current Research and Applications Caister Academic Press ISBN 978 1 904455 96 7 a b O Toole GA Kolter R May 1998 Initiation of biofilm formation in Pseudomonas fluorescens WCS365 proceeds via multiple convergent signalling pathways a genetic analysis Molecular Microbiology 28 3 449 61 doi 10 1046 j 1365 2958 1998 00797 x PMID 9632250 S2CID 43897816 O Toole GA Kolter R October 1998 Flagellar and twitching motility are necessary for Pseudomonas aeruginosa biofilm development Molecular Microbiology 30 2 295 304 doi 10 1046 j 1365 2958 1998 01062 x PMID 9791175 S2CID 25140899 Karatan E Watnick P June 2009 Signals regulatory networks and materials that build and break bacterial biofilms Microbiology and Molecular Biology Reviews 73 2 310 47 doi 10 1128 MMBR 00041 08 PMC 2698413 PMID 19487730 Hoffman LR D Argenio DA MacCoss MJ Zhang Z Jones RA Miller SI August 2005 Aminoglycoside antibiotics induce bacterial biofilm formation Nature 436 7054 1171 5 Bibcode 2005Natur 436 1171H doi 10 1038 nature03912 PMID 16121184 S2CID 4404961 primary source An D Parsek MR June 2007 The promise and peril of transcriptional profiling in biofilm communities Current Opinion in Microbiology 10 3 292 6 doi 10 1016 j mib 2007 05 011 PMID 17573234 a b Momeni B June 2018 Division of Labor How Microbes Split Their Responsibility Current Biology 28 12 R697 R699 doi 10 1016 j cub 2018 05 024 PMID 29920261 S2CID 49315067 Case C Funke B Tortora G Microbiology An Introduction tenth ed Briandet R Herry J Bellon Fontaine M August 2001 Determination of the van der Waals electron donor and electron acceptor surface tension components of static Gram positive microbial biofilms Colloids Surf B Biointerfaces 21 4 299 310 doi 10 1016 S0927 7765 00 00213 7 PMID 11397632 Takahashi H Suda T Tanaka Y Kimura B June 2010 Cellular hydrophobicity of Listeria monocytogenes involves initial attachment and biofilm formation on the surface of polyvinyl chloride Lett Appl Microbiol 50 6 618 25 doi 10 1111 j 1472 765X 2010 02842 x PMID 20438621 S2CID 24880220 7 Archaea Biology LibreTexts 6 February 2018 Madigan M 2019 Brock biology of microorganisms Fifteenth Global ed Pearson p 86 ISBN 9781292235103 a b Wang F Cvirkaite Krupovic V Krupovic M Egelman EH June 2022 Archaeal bundling pili of Pyrobaculum calidifontis reveal similarities between archaeal and bacterial biofilms Proceedings of the National Academy of Sciences of the United States of America 119 26 e2207037119 Bibcode 2022PNAS 11907037W doi 10 1073 pnas 2207037119 PMC 9245690 PMID 35727984 Golden Dome Cave National Park Service November 6 2021 Retrieved February 11 2024 a b c Donlan RM 2002 Biofilms Microbial Life on Surfaces Emerging Infectious Diseases 8 9 881 890 doi 10 3201 eid0809 020063 PMC 2732559 PMID 12194761 Li S Liu SY Chan SY Chua SL January 2022 Biofilm matrix cloaks bacterial quorum sensing chemoattractants from predator detection The ISME Journal 16 5 1388 1396 doi 10 1038 s41396 022 01190 2 PMC 9038794 PMID 35034106 Ciofu O Tolker Nielsen T 2019 Tolerance and Resistance of Pseudomonas aeruginosa Biofilms to Antimicrobial Agents How P aeruginosa Can Escape Antibiotics Frontiers in Microbiology 10 913 doi 10 3389 fmicb 2019 00913 PMC 6509751 PMID 31130925 Sakuragi Y Kolter R July 2007 Quorum sensing regulation of the biofilm matrix genes pel of Pseudomonas aeruginosa Journal of Bacteriology 189 14 5383 6 doi 10 1128 JB 00137 07 PMC 1951888 PMID 17496081 a b Rapacka Zdonczyk A Wozniak A Nakonieczna J Grinholc M February 2021 Development of Antimicrobial Phototreatment Tolerance Why the Methodology Matters International Journal of Molecular Sciences MDPI AG 22 4 2224 doi 10 3390 ijms22042224 PMC 7926562 PMID 33672375 nbsp Material was copied from this source which is available under a Creative Commons Attribution 4 0 International License Hall CW Mah TF May 2017 Molecular mechanisms of biofilm based antibiotic resistance and tolerance in pathogenic bacteria FEMS Microbiology Reviews Oxford University Press OUP 41 3 276 301 doi 10 1093 femsre fux010 PMID 28369412 O Toole G Kaplan HB Kolter R 2000 Biofilm formation as microbial development Annual Review of Microbiology 54 49 79 doi 10 1146 annurev micro 54 1 49 PMID 11018124 a b Monroe D November 2007 Looking for chinks in the armor of bacterial biofilms PLOS Biology 5 11 e307 doi 10 1371 journal pbio 0050307 PMC 2071939 PMID 18001153 Kaplan JB Ragunath C Ramasubbu N Fine DH August 2003 Detachment of Actinobacillus actinomycetemcomitans biofilm cells by an endogenous beta hexosaminidase activity Journal of Bacteriology 185 16 4693 8 doi 10 1128 JB 185 16 4693 4698 2003 PMC 166467 PMID 12896987 Izano EA Amarante MA Kher WB Kaplan JB January 2008 Differential roles of poly N acetylglucosamine surface polysaccharide and extracellular DNA in Staphylococcus aureus and Staphylococcus epidermidis biofilms Applied and Environmental Microbiology 74 2 470 6 Bibcode 2008ApEnM 74 470I doi 10 1128 AEM 02073 07 PMC 2223269 PMID 18039822 Kaplan JB Ragunath C Velliyagounder K Fine DH Ramasubbu N July 2004 Enzymatic detachment of Staphylococcus epidermidis biofilms Antimicrobial Agents and Chemotherapy 48 7 2633 6 doi 10 1128 AAC 48 7 2633 2636 2004 PMC 434209 PMID 15215120 Xavier JB Picioreanu C Rani SA van Loosdrecht MC Stewart PS December 2005 Biofilm control strategies based on enzymic disruption of the extracellular polymeric substance matrix a modelling study Microbiology 151 Pt 12 3817 32 doi 10 1099 mic 0 28165 0 PMID 16339929 Davies DG Marques CN March 2009 A fatty acid messenger is responsible for inducing dispersion in microbial biofilms Journal of Bacteriology 191 5 1393 403 doi 10 1128 JB 01214 08 PMC 2648214 PMID 19074399 Barraud N Hassett DJ Hwang SH Rice SA Kjelleberg S Webb JS 2006 Involvement of nitric oxide in biofilm dispersal of Pseudomonas aeruginosa Journal of Bacteriology 188 21 7344 7353 doi 10 1128 jb 00779 06 PMC 1636254 PMID 17050922 Barraud N Storey MV Moore ZP Webb JS Rice SA Kjelleberg S 2009 Nitric oxide mediated dispersal in single and multi species biofilms of clinically and industrially relevant microorganisms Microbial Biotechnology 2 3 370 378 doi 10 1111 j 1751 7915 2009 00098 x PMC 3815757 PMID 21261931 Dispersal of Biofilm in Cystic Fibrosis using Low Dose Nitric Oxide University of Southampton Retrieved 20 January 2012 a b Chua SL Liu Y Yam JK Tolker Nielsen T Kjelleberg S Givskov M Yang L 2014 Dispersed cells represent a distinct stage in the transition from bacterial biofilm to planktonic lifestyles Nature Communications 5 4462 Bibcode 2014NatCo 5 4462C doi 10 1038 ncomms5462 PMID 25042103 Chua SL Hultqvist LD Yuan M Rybtke M Nielsen TE Givskov M Tolker Nielsen T Yang L August 2015 In vitro and in vivo generation and characterization of Pseudomonas aeruginosa biofilm dispersed cells via c di GMP manipulation Nat Protoc 10 8 1165 80 doi 10 1038 nprot 2015 067 hdl 10356 84100 PMID 26158442 S2CID 20235088 a b Ma Y Deng Y Hua H Khoo BL Chua SL August 2023 Distinct bacterial population dynamics and disease dissemination after biofilm dispersal and disassembly The ISME Journal 17 8 1290 1302 doi 10 1038 s41396 023 01446 5 PMC 10356768 PMID 37270584 Nadell CD Xavier JB Foster KR January 2009 The sociobiology of biofilms FEMS Microbiology Reviews 33 1 206 24 doi 10 1111 j 1574 6976 2008 00150 x PMID 19067751 a b c d Rybtke M Hultqvist LD Givskov M Tolker Nielsen T November 2015 Pseudomonas aeruginosa Biofilm Infections Community Structure Antimicrobial Tolerance and Immune Response Journal of Molecular Biology 427 23 3628 45 doi 10 1016 j jmb 2015 08 016 PMID 26319792 Danese PN Pratt LA Kolter R June 2000 Exopolysaccharide production is required for development of Escherichia coli K 12 biofilm architecture Journal of Bacteriology 182 12 3593 6 doi 10 1128 jb 182 12 3593 3596 2000 PMC 101973 PMID 10852895 Branda SS Chu F Kearns DB Losick R Kolter R February 2006 A major protein component of the Bacillus subtilis biofilm matrix Molecular Microbiology 59 4 1229 38 doi 10 1111 j 1365 2958 2005 05020 x PMID 16430696 S2CID 3041295 Choong FX Back M Fahlen S Johansson LB Melican K Rhen M et al 23 November 2016 Salmonella biofilms using luminescent oligothiophenes npj Biofilms and Microbiomes 2 16024 doi 10 1038 npjbiofilms 2016 24 PMC 5515270 PMID 28721253 Flemming HC Wingender J Szewzyk U Steinberg P Rice SA Kjelleberg S August 2016 Biofilms an emergent form of bacterial life Nature Reviews Microbiology 14 9 563 75 doi 10 1038 nrmicro 2016 94 PMID 27510863 S2CID 4384131 Stoodley P Debeer D Lewandowski Z August 1994 Liquid flow in biofilm systems Applied and Environmental Microbiology 60 8 2711 6 Bibcode 1994ApEnM 60 2711S doi 10 1128 aem 60 8 2711 2716 1994 PMC 201713 PMID 16349345 Vlamakis H Aguilar C Losick R Kolter R April 2008 Control of cell fate by the formation of an architecturally complex bacterial community Genes amp Development 22 7 945 53 doi 10 1101 gad 1645008 PMC 2279205 PMID 18381896 Stewart PS Costerton JW July 2001 Antibiotic resistance of bacteria in biofilms Lancet 358 9276 135 8 doi 10 1016 S0140 6736 01 05321 1 PMID 11463434 S2CID 46125592 Pandey R Mishra SK Shrestha A 2021 Characterisation of ESKAPE Pathogens with Special Reference to Multidrug Resistance and Biofilm Production in a Nepalese Hospital Infect Drug Resist 14 2201 2212 doi 10 2147 IDR S306688 PMC 8214009 PMID 34163185 a b c d e f Del Pozo JL Rouse MS Patel R September 2008 Bioelectric effect and bacterial biofilms A systematic review The International Journal of Artificial Organs 31 9 786 795 doi 10 1177 039139880803100906 PMC 3910516 PMID 18924090 a b Chimileski S Franklin MJ Papke RT August 2014 Biofilms formed by the archaeon Haloferax volcanii exhibit cellular differentiation and social motility and facilitate horizontal gene transfer BMC Biology 12 65 doi 10 1186 s12915 014 0065 5 PMC 4180959 PMID 25124934 Molin S Tolker Nielsen T June 2003 Gene transfer occurs with enhanced efficiency in biofilms and induces enhanced stabilisation of the biofilm structure Current Opinion in Biotechnology 14 3 255 61 doi 10 1016 S0958 1669 03 00036 3 PMID 12849777 Jakubovics NS Shields RC Rajarajan N Burgess JG December 2013 Life after death the critical role of extracellular DNA in microbial biofilms Letters in Applied Microbiology 57 6 467 75 doi 10 1111 lam 12134 PMID 23848166 S2CID 206168952 Spoering AL Lewis K December 2001 Biofilms and planktonic cells of Pseudomonas aeruginosa have similar resistance to killing by antimicrobials Journal of Bacteriology 183 23 6746 51 doi 10 1128 JB 183 23 6746 6751 2001 PMC 95513 PMID 11698361 Introduction to Biofilms Desirable and undesirable impacts of biofilm Archived from the original on 22 June 2008 primary source Andersen PC Brodbeck BV Oden S Shriner A Leite B September 2007 Influence of xylem fluid chemistry on planktonic growth biofilm formation and aggregation of Xylella fastidiosa FEMS Microbiology Letters 274 2 210 7 doi 10 1111 j 1574 6968 2007 00827 x PMID 17610515 Biological wastewater treatment processes secondary treatment Staffordshire University Archived from the original on 18 April 2011 Retrieved 13 December 2019 Centre for Affordable Water and Sanitation Technology Biosand Filter Manual Design Construction amp Installation July 2007 Slow Sand Filtration PDF Tech Brief Morgantown WV National Drinking Water Clearinghouse U S 14 June 2000 Archived from the original PDF on 6 April 2016 Kloepper JW 1988 Plant Growth Promoting Rhizobacteria on Canola Rapeseed Plant Disease 72 1 42 doi 10 1094 pd 72 0042 ISSN 0191 2917 a b c d Nihorimbere V Cawoy H Seyer A Brunelle A Thonart P Ongena M January 2012 Impact of rhizosphere factors on cyclic lipopeptide signature from the plant beneficial strain Bacillus amyloliquefaciens S499 FEMS Microbiology Ecology 79 1 176 91 Bibcode 2012FEMME 79 176N doi 10 1111 j 1574 6941 2011 01208 x PMID 22029651 Choudhary DK Johri BN September 2009 Interactions of Bacillus spp and plants with special reference to induced systemic resistance ISR Microbiological Research 164 5 493 513 doi 10 1016 j micres 2008 08 007 PMID 18845426 a b van Loon LC 5 June 2007 Plant responses to plant growth promoting rhizobacteria European Journal of Plant Pathology 119 3 243 254 doi 10 1007 s10658 007 9165 1 ISSN 0929 1873 a b c d e Van Wees SC Van der Ent S Pieterse CM August 2008 Plant immune responses triggered by beneficial microbes Current Opinion in Plant Biology 11 4 443 8 doi 10 1016 j pbi 2008 05 005 hdl 1874 30010 PMID 18585955 S2CID 25880745 Holguin G Bashan Y December 1996 Nitrogen fixation by Azospirillum brasilense Cd is promoted when co cultured with a mangrove rhizosphere bacterium Staphylococcus sp Soil Biology and Biochemistry 28 12 1651 1660 doi 10 1016 s0038 0717 96 00251 9 ISSN 0038 0717 Babalola OO November 2010 Beneficial bacteria of agricultural importance Biotechnology Letters 32 11 1559 70 doi 10 1007 s10529 010 0347 0 PMID 20635120 S2CID 13518392 Bakker PA Pieterse CM van Loon LC February 2007 Induced Systemic Resistance by Fluorescent Pseudomonas spp Phytopathology 97 2 239 43 doi 10 1094 phyto 97 2 0239 PMID 18944381 Bent E 2006 Induced Systemic Resistance Mediated by Plant Growth Promoting Rhizobacteria PGPR and Fungi PGPF Multigenic and Induced Systemic Resistance in Plants Springer US pp 225 258 doi 10 1007 0 387 23266 4 10 ISBN 9780387232652 Lynch JM Brimecombe MJ De Leij FA 21 August 2001 Rhizosphere eLS John Wiley amp Sons Ltd doi 10 1038 npg els 0000403 ISBN 0470016175 Randal Bollinger R Barbas AS Bush EL Lin SS Parker W December 2007 Biofilms in the large bowel suggest an apparent function of the human vermiform appendix Journal of Theoretical Biology 249 4 826 31 Bibcode 2007JThBi 249 826R doi 10 1016 j jtbi 2007 08 032 PMID 17936308 Buret AG Motta JP Allain T Ferraz J Wallace JL January 2019 Pathobiont release from dysbiotic gut microbiota biofilms in intestinal inflammatory diseases a role for iron Journal of Biomedical Science 26 1 1 doi 10 1186 s12929 018 0495 4 PMC 6317250 PMID 30602371 Characklis WG Nevimons MJ Picologlou BF 1981 Influence of Fouling Biofilms on Heat Transfer Heat Transfer Engineering 3 1 23 37 Bibcode 1981HTrEn 3 23C doi 10 1080 01457638108939572 Schwermer CU Lavik G Abed RM Dunsmore B Ferdelman TG Stoodley P et al May 2008 Impact of nitrate on the structure and function of bacterial biofilm communities in pipelines used for injection of seawater into oil fields Applied and Environmental Microbiology 74 9 2841 51 Bibcode 2008ApEnM 74 2841S doi 10 1128 AEM 02027 07 PMC 2394879 PMID 18344353 Chandki R Banthia P Banthia R April 2011 Biofilms A microbial home Journal of Indian Society of Periodontology 15 2 111 4 doi 10 4103 0972 124X 84377 PMC 3183659 PMID 21976832 Augustin M Chifiriuc CB Lazăr V Stănescu R Burlibașa M Ispas DC December 2010 Microbial biofilms in dental medicine in reference to implanto prostethic rehabilitation Revista de Chirurgie Oro maxilo facială și Implantologie in Romanian 1 1 9 13 ISSN 2069 3850 8 Retrieved 3 June 2012 permanent dead link webpage has a translation button Marquis RE September 1995 Oxygen metabolism oxidative stress and acid base physiology of dental plaque biofilms Journal of Industrial Microbiology 15 3 198 207 doi 10 1007 bf01569826 PMID 8519478 S2CID 19959528 a b c Lemos JA Abranches J Burne RA January 2005 Responses of cariogenic streptococci to environmental stresses PDF Current Issues in Molecular Biology 7 1 95 107 PMID 15580782 Archived from the original PDF on 7 April 2014 Retrieved 3 April 2014 Tamm C Hodes ME Chargaff E March 1952 The formation apurinic acid from the desoxyribonucleic acid of calf thymus The Journal of Biological Chemistry 195 1 49 63 doi 10 1016 S0021 9258 19 50874 2 PMID 14938354 Freese EB April 1961 Transitions and transversions induced by depurinating agents Proceedings of the National Academy of Sciences of the United States of America 47 4 540 5 Bibcode 1961PNAS 47 540B doi 10 1073 pnas 47 4 540 PMC 221484 PMID 13701660 a b Pennwell Toothbrush technology dentifrices and dental biofilm removal Dental Academy of CE Accessed 12 January 2022 Fejerskov O 2015 Pathology of dental caries In Dental caries the disease and its clinical management Oxford UK Wiley Blackwell pp 7 9 ISBN 978 1405138895 a b Li YH Lau PC Lee JH Ellen RP Cvitkovitch DG February 2001 Natural genetic transformation of Streptococcus mutans growing in biofilms J Bacteriol 183 3 897 908 doi 10 1128 JB 183 3 897 908 2001 PMC 94956 PMID 11208787 Senadheera D Cvitkovitch DG 2008 Quorum Sensing and Biofilm Formation by Streptococcus mutans Bacterial Signal Transduction Networks and Drug Targets Advances in Experimental Medicine and Biology Vol 631 pp 178 88 doi 10 1007 978 0 387 78885 2 12 ISBN 978 0 387 78884 5 PMID 18792689 a b Michod RE Bernstein H Nedelcu AM May 2008 Adaptive value of sex in microbial pathogens Infect Genet Evol 8 3 267 85 doi 10 1016 j meegid 2008 01 002 PMID 18295550 http www hummingbirds arizona edu Faculty Michod Downloads IGE 20review 20sex pdf Atkinson S Goldstone RJ Joshua GW Chang CY Patrick HL Camara M et al January 2011 Biofilm development on Caenorhabditis elegans by Yersinia is facilitated by quorum sensing dependent repression of type III secretion PLOS Pathogens 7 1 e1001250 doi 10 1371 journal ppat 1001250 PMC 3017118 PMID 21253572 Chan SY Liu SY Seng Z Chua SL September 2020 Biofilm matrix disrupts nematode motility and predatory behavior The ISME Journal 15 1 260 269 doi 10 1038 s41396 020 00779 9 PMC 7852553 PMID 32958848 Li S Liu SY Chan SY Chua SL May 2022 Biofilm matrix cloaks bacterial quorum sensing chemoattractants from predator detection The ISME Journal 16 5 1388 1396 doi 10 1038 s41396 022 01190 2 PMC 9038794 PMID 35034106 Abee T Kovacs AT Kuipers OP van der Veen S April 2011 Biofilm formation and dispersal in Gram positive bacteria PDF Current Opinion in Biotechnology 22 2 172 9 doi 10 1016 j copbio 2010 10 016 hdl 11370 999da2a4 d509 471b bab5 085dac6ff681 PMID 21109420 S2CID 22024410 Rossi F De Philippis R April 2015 Role of cyanobacterial exopolysaccharides in phototrophic biofilms and in complex microbial mats Life 5 2 1218 38 Bibcode 2015Life 5 1218R doi 10 3390 life5021218 PMC 4500136 PMID 25837843 a b Danhorn T Fuqua C 2007 Biofilm formation by plant associated bacteria Annual Review of Microbiology 61 401 22 doi 10 1146 annurev micro 61 080706 093316 PMID 17506679 a b Joubert LM Wolfaardt GM Botha A August 2006 Microbial exopolymers link predator and prey in a model yeast biofilm system Microb Ecol 52 2 187 97 doi 10 1007 s00248 006 9063 7 PMID 16897306 S2CID 20431229 a b Van Colen C Underwood GC Serodio J Paterson DM 2014 Ecology of intertidal microbial biofilms Mechanisms patterns and future research needs Journal of Sea Research 92 2 5 Bibcode 2014JSR 92 2V doi 10 1016 j seares 2014 07 003 a b Aslam SN Cresswell Maynard T Thomas DN Underwood GJ December 2012 Production and Characterization of the Intra and Extracellular Carbohydrates and Polymeric Substances EPS of Three Sea Ice Diatom Species and Evidence for a Cryoprotective Role for EPS J Phycol 48 6 1494 509 doi 10 1111 jpy 12004 PMID 27009999 S2CID 9226690 Research on microbial biofilms PA 03 047 NIH National Heart Lung and Blood Institute 20 December 2002 Rogers A 2008 Molecular Oral Microbiology Caister Academic Press pp 88 91 ISBN 978 1 904455 24 0 Imamura Y Chandra J Mukherjee PK Lattif AA Szczotka Flynn LB Pearlman E et al January 2008 Fusarium and Candida albicans biofilms on soft contact lenses model development influence of lens type and susceptibility to lens care solutions Antimicrobial Agents and Chemotherapy 52 1 171 82 doi 10 1128 AAC 00387 07 PMC 2223913 PMID 17999966 Capoor MN Ruzicka F Schmitz JE James GA Machackova T Jancalek R et al 3 April 2017 Propionibacterium acnes biofilm is present in intervertebral discs of patients undergoing microdiscectomy PLOS ONE 12 4 e0174518 Bibcode 2017PLoSO 1274518C doi 10 1371 journal pone 0174518 PMC 5378350 PMID 28369127 Lewis K April 2001 Riddle of biofilm resistance Antimicrobial Agents and Chemotherapy 45 4 999 1007 doi 10 1128 AAC 45 4 999 1007 2001 PMC 90417 PMID 11257008 Parsek MR Singh PK 2003 Bacterial biofilms an emerging link to disease pathogenesis Annual Review of Microbiology 57 677 701 doi 10 1146 annurev micro 57 030502 090720 PMID 14527295 a b Agarwal A Mooney M Agarwal AG Jayaswal D Saakyan G Goel V et al 2020 High Prevalence of Biofilms on Retrieved Implants from Aseptic Pseudarthrosis Cases Spine Surgery and Related Research 5 2 104 108 doi 10 22603 ssrr 2020 0147 PMC 8026210 PMID 33842718 a b Curran N 20 November 2020 New study first to visually capture biofilm architecture in retrieved implants from live patients Spinal News International Retrieved 24 November 2020 biofilm 22 December 2020 Davis SC Ricotti C Cazzaniga A Welsh E Eaglstein WH Mertz PM 2008 Microscopic and physiologic evidence for biofilm associated wound colonization in vivo Wound Repair and Regeneration 16 1 23 9 doi 10 1111 j 1524 475X 2007 00303 x PMID 18211576 S2CID 205669081 Vyas KS Wong LK January 2016 Detection of Biofilm in Wounds as an Early Indicator for Risk for Tissue Infection and Wound Chronicity Annals of Plastic Surgery 76 1 127 31 doi 10 1097 SAP 0000000000000440 PMID 25774966 S2CID 42078581 Sanclement J Webster P Thomas J Ramadan H 2005 Bacterial biofilms in surgical specimens of patients with chronic rhinosinusitis The Laryngoscope 115 4 578 82 doi 10 1097 01 mlg 0000161346 30752 18 PMID 15805862 S2CID 25830188 Sanderson AR Leid JG Hunsaker D July 2006 Bacterial biofilms on the sinus mucosa of human subjects with chronic rhinosinusitis The Laryngoscope 116 7 1121 6 doi 10 1097 01 mlg 0000221954 05467 54 PMID 16826045 S2CID 24785016 Leevy WM Gammon ST Jiang H Johnson JR Maxwell DJ Jackson EN et al December 2006 Optical imaging of bacterial infection in living mice using a fluorescent near infrared molecular probe Journal of the American Chemical Society 128 51 16476 7 doi 10 1021 ja0665592 PMC 2531239 PMID 17177377 Kaplan JB Izano EA Gopal P Karwacki MT Kim S Bose JL et al 2012 Low levels of b lactam antibiotics induce extracellular DNA release and biofilm formation in Staphylococcus aureus mBio 3 4 e00198 12 doi 10 1128 mBio 00198 12 PMC 3419523 PMID 22851659 Ibrahim AM 2015 The tragedy of the commons and prisoner s dilemma may improve our realization of the theory of life and provide us with advanced therapeutic ways Report doi 10 13140 RG 2 1 2327 9842 Ciofu O Tolker Nielsen T Jensen PO Wang H Hoiby N May 2015 Antimicrobial resistance respiratory tract infections and role of biofilms in lung infections in cystic fibrosis patients Advanced Drug Delivery Reviews 85 7 23 doi 10 1016 j addr 2014 11 017 PMID 25477303 Whitchurch CB Tolker Nielsen T Ragas PC Mattick JS February 2002 Extracellular DNA required for bacterial biofilm formation Science 295 5559 1487 doi 10 1126 science 295 5559 1487 PMID 11859186 Sen CK Gordillo GM Roy S Kirsner R Lambert L Hunt TK et al November 2009 Human skin wounds a major and snowballing threat to public health and the economy Wound Repair and Regeneration 17 6 763 771 doi 10 1111 j 1524 475X 2009 00543 x PMC 2810192 PMID 19903300 Singh PK Parsek MR Greenberg EP Welsh MJ May 2002 A component of innate immunity prevents bacterial biofilm development Nature 417 6888 552 555 Bibcode 2002Natur 417 552S doi 10 1038 417552a PMID 12037568 S2CID 4423528 Oggioni MR Trappetti C Kadioglu A Cassone M Iannelli F Ricci S et al September 2006 Switch from planktonic to sessile life a major event in pneumococcal pathogenesis Molecular Microbiology 61 5 1196 210 doi 10 1111 j 1365 2958 2006 05310 x PMC 1618759 PMID 16925554 Wei H Havarstein LS August 2012 Fratricide is essential for efficient gene transfer between pneumococci in biofilms Appl Environ Microbiol 78 16 5897 905 Bibcode 2012ApEnM 78 5897W doi 10 1128 AEM 01343 12 PMC 3406168 PMID 22706053 Kalsy M Tonk M Hardt M Dobrindt U Zdybicka Barabas A Cytrynska M Vilcinskas A Mukherjee K 2020 The insect antimicrobial peptide cecropin A disrupts uropathogenic Escherichia coli biofilms npj Biofilms and Microbiomes 6 1 6 doi 10 1038 s41522 020 0116 3 PMC 7016129 PMID 32051417 Sturbelle RT de Avila LF Roos TB Borchardt JL da Conceicao R Dellagostin OA Leite FP November 2015 The role of quorum sensing in Escherichia coli ETEC virulence factors Veterinary Microbiology 180 3 4 245 252 doi 10 1016 j vetmic 2015 08 015 PMID 26386492 Vogeleer P Tremblay YD Mafu AA Jacques M Harel J 2014 Life on the outside role of biofilms in environmental persistence of Shiga toxin producing Escherichia coli Frontiers in Microbiology 5 317 doi 10 3389 fmicb 2014 00317 PMC 4076661 PMID 25071733 Danese PN Pratt LA Kolter R June 2000 Exopolysaccharide production is required for development of Escherichia coli K 12 biofilm architecture Journal of Bacteriology 182 12 3593 3596 doi 10 1128 JB 182 12 3593 3596 2000 PMC 101973 PMID 10852895 Niranjan V Malini A June 2014 Antimicrobial resistance pattern in Escherichia coli causing urinary tract infection among inpatients The Indian Journal of Medical Research 139 6 945 948 PMC 4165009 PMID 25109731 Reisner A Maierl M Jorger M Krause R Berger D Haid A et al March 2014 Type 1 fimbriae contribute to catheter associated urinary tract infections caused by Escherichia coli Journal of Bacteriology 196 5 931 939 doi 10 1128 JB 00985 13 PMC 3957706 PMID 24336940 Kobayashi SD Malachowa N Whitney AR Braughton KR Gardner DJ Long D et al September 2011 Comparative analysis of USA300 virulence determinants in a rabbit model of skin and soft tissue infection The Journal of Infectious Diseases 204 6 937 941 doi 10 1093 infdis jir441 PMC 3156927 PMID 21849291 Kitur K Parker D Nieto P Ahn DS Cohen TS Chung S et al April 2015 Toxin induced necroptosis is a major mechanism of Staphylococcus aureus lung damage PLOS Pathogens 11 4 e1004820 doi 10 1371 journal ppat 1004820 PMC 4399879 PMID 25880560 Thurlow LR Hanke ML Fritz T Angle A Aldrich A Williams SH et al June 2011 Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo Journal of Immunology 186 11 6585 6596 doi 10 4049 jimmunol 1002794 PMC 3110737 PMID 21525381 Craft KM Nguyen JM Berg LJ Townsend SD August 2019 Methicillin resistant Staphylococcus aureus MRSA antibiotic resistance and the biofilm phenotype MedChemComm 10 8 1231 1241 doi 10 1039 c9md00044e PMC 6748282 PMID 31534648 Lazar V December 2011 Quorum sensing in biofilms how to destroy the bacterial citadels or their cohesion power Anaerobe 17 6 280 5 doi 10 1016 j anaerobe 2011 03 023 PMID 21497662 Bjarnsholt T Jensen PO Moser C Hoiby N 2011 Biofilm infections New York Springer ISBN 9781441960832 OCLC 682907381 a b c d Bryers JD May 2008 Medical biofilms Biotechnology and Bioengineering 100 1 1 18 doi 10 1002 bit 21838 PMC 2706312 PMID 18366134 Auler ME Morreira D Rodrigues FF Abr Ao MS Margarido PF Matsumoto FE et al February 2010 Biofilm formation on intrauterine devices in patients with recurrent vulvovaginal candidiasis Medical Mycology 48 1 211 6 doi 10 3109 13693780902856626 PMID 20055746 Vuotto C Longo F Balice MP Donelli G Varaldo PE September 2014 Antibiotic Resistance Related to Biofilm Formation in Klebsiella pneumoniae Pathogens 3 3 743 758 doi 10 3390 pathogens3030743 PMC 4243439 PMID 25438022 Santos Lopez A Marshall CW Scribner MR Snyder DJ Cooper VS September 2019 Evolutionary pathways to antibiotic resistance are dependent upon environmental structure and bacterial lifestyle eLife 8 e47612 doi 10 7554 eLife 47612 PMC 6814407 PMID 31516122 Pai L Patil S Liu S Wen F 2023 A growing battlefield in the war against biofilm induced antimicrobial resistance insights from reviews on antibiotic resistance Front Cell Infect Microbiol 13 1327069 doi 10 3389 fcimb 2023 1327069 PMC 10770264 PMID 38188636 a b Kim YW Subramanian S Gerasopoulos K Ben Yoav H Wu HC Quan D et al 2015 Effect of electrical energy on the efficacy of biofilm treatment using the bioelectric effect npj Biofilms and Microbiomes 1 15016 doi 10 1038 npjbiofilms 2015 16 PMC 5515217 PMID 28721233 Martins dos Santos VA Yakimov MM Timmis KN Golyshin PN 2008 Genomic Insights into Oil Biodegradation in Marine Systems In Diaz E ed Microbial Biodegradation Genomics and Molecular Biology Horizon Scientific Press pp 1971 ISBN 978 1 904455 17 2 Wang VB Chua SL Cai Z Sivakumar K Zhang Q Kjelleberg S et al March 2014 A stable synergistic microbial consortium for simultaneous azo dye removal and bioelectricity generation Bioresource Technology 155 71 76 doi 10 1016 j biortech 2013 12 078 PMID 24434696 Wang VB Chua SL Cao B Seviour T Nesatyy VJ Marsili E et al 2013 Engineering PQS biosynthesis pathway for enhancement of bioelectricity production in pseudomonas aeruginosa microbial fuel cells PLOS ONE 8 5 e63129 Bibcode 2013PLoSO 863129W doi 10 1371 journal pone 0063129 PMC 3659106 PMID 23700414 Vera M Schippers A Sand W September 2013 Progress in bioleaching fundamentals and mechanisms of bacterial metal sulfide oxidation part A Appl Microbiol Biotechnol 97 17 7529 41 doi 10 1007 s00253 013 4954 2 PMID 23720034 S2CID 17677624 Chan SY Wong MW Kwan BT Fang JK Chua SL 12 October 2022 Microbial Enzymatic Combinatorial Approach to Capture and Release Microplastics Environmental Science amp Technology Letters 9 11 975 982 Bibcode 2022EnSTL 9 975C doi 10 1021 acs estlett 2c00558 ISSN 2328 8930 S2CID 252892619 Liu SY Leung MM Fang JK Chua SL 15 January 2021 Engineering a microbial trap and release mechanism for microplastics removal Chemical Engineering Journal 404 127079 doi 10 1016 j cej 2020 127079 hdl 10397 88307 ISSN 1385 8947 S2CID 224972583 a b c d e f g h i Srey S Jahid ID Ha SD June 2013 Biofilm formation in food industries A food safety concern Food Control 31 2 572 585 doi 10 1016 j foodcont 2012 12 001 ISSN 0956 7135 T Tarver Biofilms A Threat to Food Safety IFT org Ift org 2016 a b c d Kumar CG Anand SK June 1998 Significance of microbial biofilms in food industry a review International Journal of Food Microbiology 42 1 2 9 27 doi 10 1016 s0168 1605 98 00060 9 PMID 9706794 Kwok TY Ma Y Chua SL April 2022 Biofilm dispersal induced by mechanical cutting leads to heightened foodborne pathogen dissemination Food Microbiology 102 103914 doi 10 1016 j fm 2021 103914 hdl 10397 100037 PMID 34809940 S2CID 244234814 a b c d Mizan F 2015 Microbial biofilms in seafood A food hygiene challenge Food Microbiology 49 41 55 doi 10 1016 j fm 2015 01 009 PMID 25846914 De Araujo LV Abreu F Lins U Santa Anna LM Nitschke M Freire DM January 2011 Rhamnolipid and surfactin inhibit Listeria monocytogenes adhesion Food Research International 44 1 481 488 doi 10 1016 j foodres 2010 09 002 Wang X Yao X Zhu Z Tang T Dai K Sadovskaya I et al July 2009 Effect of berberine on Staphylococcus epidermidis biofilm formation International Journal of Antimicrobial Agents 34 1 60 6 doi 10 1016 j ijantimicag 2008 10 033 PMID 19157797 Carvalho DB Fox EG Santos DG Sousa JS Freire DM Nogueira FC et al July 2019 Fire Ant Venom Alkaloids Inhibit Biofilm Formation Toxins 11 7 420 doi 10 3390 toxins11070420 PMC 6669452 PMID 31323790 Braithwaite RA McEvoy LA 2004 Marine biofouling on fish farms and its remediation Advances in Marine Biology 47 215 252 doi 10 1016 S0065 2881 04 47003 5 ISBN 9780120261482 PMID 15596168 Qian PY Lau SC Dahms HU Dobretsov S Harder T 2007 Marine biofilms as mediators of colonization by marine macroorganisms implications for antifouling and aquaculture Marine Biotechnology 9 4 399 410 doi 10 1007 s10126 007 9001 9 PMID 17497196 S2CID 7614961 Cai W De La Fuente L Arias CR September 2013 Biofilm formation by the fish pathogen Flavobacterium columnare development and parameters affecting surface attachment Applied and Environmental Microbiology 79 18 5633 42 Bibcode 2013ApEnM 79 5633C doi 10 1128 AEM 01192 13 PMC 3754160 PMID 23851087 King RK Flick Jr GJ Pierson D Smith SA Boardman GD Coale Jr CW 2004 Identification of bacterial pathogens in biofilms of recirculating aquaculture systems Journal of Aquatic Food Product Technology 13 125 133 doi 10 1300 j030v13n01 11 S2CID 83791439 Bourne DG Hoj L Webster NS Swan J Hall MR 2006 Biofilm development within a larval rearing tank of the tropical rock lobster Panulirus ornatus Aquaculture 260 1 4 27 38 doi 10 1016 j aquaculture 2006 06 023 Wietz M Hall MR Hoj L July 2009 Effects of seawater ozonation on biofilm development in aquaculture tanks Systematic and Applied Microbiology 32 4 266 77 doi 10 1016 j syapm 2009 04 001 PMID 19446976 Liu YS Deng Y Chen CK Khoo BL Chua SL June 2022 Rapid detection of microorganisms in a fish infection microfluidics platform Journal of Hazardous Materials 431 128572 doi 10 1016 j jhazmat 2022 128572 PMID 35278965 S2CID 247136872 Karunasagar I Pai R Malathi G 1994 Mass mortality of Penaeus monodon larvae due to antibiotic resistant Vibrio harveyi infection Aquaculture 128 3 4 203 209 doi 10 1016 0044 8486 94 90309 3 Lawrence JR Korber DR Hoyle BD Costerton JW Caldwell DE October 1991 Optical sectioning of microbial biofilms Journal of Bacteriology 173 20 6558 67 doi 10 1128 jb 173 20 6558 6567 1991 PMC 208993 PMID 1917879 Cooksey K Wigglesworth Cooksey B 1995 Adhesion of bacteria and diatoms to surfaces in the sea a review Aquatic Microbial Ecology 9 1 87 96 doi 10 3354 ame009087 Fanning S Mitchell AP 2012 Fungal Biofilms PLOS Pathog 8 4 e1002585 doi 10 1371 journal ppat 1002585 PMC 3320593 PMID 22496639 Chandra J Kuhn DM Mukherjee PK Hoyer LL McCormick T Ghannoum MA September 2001 Biofilm formation by the fungal pathogen Candida albicans development architecture and drug resistance J Bacteriol 183 18 5385 94 doi 10 1128 jb 183 18 5385 5394 2001 PMC 95423 PMID 11514524 Burmolle M Kjoller A Sorenses S 2012 Lear G Gavin L Lewis G eds Microbial Biofilms Current Research and Applications Horizon Scientific Press pp 61 71 ISBN 978 1904455967 Steele DJ Franklin DJ Underwood GJ September 2014 Protection of cells from salinity stress by extracellular polymeric substances in diatom biofilms Biofouling 30 8 987 98 doi 10 1080 08927014 2014 960859 PMC 4706044 PMID 25268215 Windler M Leinweber K Bartulos CR Philipp B Kroth PG April 2015 Biofilm and capsule formation of the diatom Achnanthidium minutissimum are affected by a bacterium J Phycol 51 2 343 55 Bibcode 2015JPcgy 51 343W doi 10 1111 jpy 12280 PMID 26986529 S2CID 1446573 Buhmann M Kroth PG Schleheck D February 2012 Photoautotrophic heterotrophic biofilm communities a laboratory incubator designed for growing axenic diatoms and bacteria in defined mixed species biofilms Environ Microbiol Rep 4 1 133 40 doi 10 1111 j 1758 2229 2011 00315 x PMID 23757240 Thomas CM Nielsen KM September 2005 Mechanisms of and barriers to horizontal gene transfer between bacteria Nature Reviews Microbiology 3 9 711 721 doi 10 1038 nrmicro1234 PMID 16138099 S2CID 1231127 a b c d e f g h i Luo A Wang F Sun D Liu X Xin B 2022 Formation Development and Cross Species Interactions in Biofilms Frontiers in Microbiology 12 757327 doi 10 3389 fmicb 2021 757327 PMC 8764401 PMID 35058893 Patkowski JB Dahlberg T Amin H Gahlot DK Vijayrajratnam S Vogel JP et al April 2023 The F pilus biomechanical adaptability accelerates conjugative dissemination of antimicrobial resistance and biofilm formation Nature Communications 14 1 1879 doi 10 1038 s41467 023 37600 y PMC 10076315 PMID 37019921 Thomas VC Hancock LE September 2009 Suicide and fratricide in bacterial biofilms The International Journal of Artificial Organs 32 9 537 544 doi 10 1177 039139880903200902 PMID 19851979 S2CID 201969291 a b c d e f g h i j Abe K Nomura N Suzuki S May 2020 Biofilms hot spots of horizontal gene transfer HGT in aquatic environments with a focus on a new HGT mechanism FEMS Microbiology Ecology 96 5 doi 10 1093 femsec fiaa031 PMC 7189800 PMID 32109282 Chimileski S Franklin MJ Papke RT August 2014 Biofilms formed by the archaeon Haloferax volcanii exhibit cellular differentiation and social motility and facilitate horizontal gene transfer BMC Biology 12 1 65 doi 10 1186 s12915 014 0065 5 PMC 4180959 PMID 25124934 Azeredo J Azevedo NF Briandet R Cerca N Coenye T Costa AR et al May 2017 Critical review on biofilm methods Critical Reviews in Microbiology 43 3 313 351 doi 10 1080 1040841X 2016 1208146 hdl 1822 45004 PMID 27868469 S2CID 3991858 Further reading editAllison DG 2000 Community structure and co operation in biofilms Cambridge UK Cambridge University Press ISBN 978 0 521 79302 5 Lynch JF Lappin Scott HM Costerton JW 2003 Microbial biofilms Cambridge UK Cambridge University Press ISBN 978 0 521 54212 8 Fratamico M 2009 Biofilms in the food and beverage industries Woodhead Publishing Limited ISBN 978 1 84569 477 7 External links editA TED ED animation on basic biofilm biology The microbial jungles all over the place and you by Scott Chimileski and Roberto Kolter Thickness analysis organic and mineral proportion of biofilms in order to decide a treatment strategy Biofilm Archive of Biofilm Research amp News Archived 9 October 2010 at the Wayback Machine Why Am I Still Sick The Movie 2012 Documentary on Biofilms The Silent Role of Biofilms in Chronic Disease HD Video Interviews on biofilms antibiotics etc with experts youtube com ADRSupport biofilm Retrieved from https en wikipedia org w index php title Biofilm amp oldid 1207136359, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.