Skip to main content

A narrative review on bacterial biofilm: its formation, clinical aspects and inhibition strategies

Abstract

Background

The predominant mode of life of most of the bacteria is their biofilm state. Based on the type of bacteria existing within the biofilm, it might be beneficial or harmful. Its beneficial aspects have been exploited mostly in waste management strategies. These biofilms affected the food industry, dairy industry and oil industry, causing huge losses by food spoilage, reduced heat transfer efficiencies and corrosion caused by biofilms in pipelines. They were considered a crucial risk to human well-being. Biofilms were responsible for more than 75% of the clinical infections caused in humans.

The main body of the abstract

Biofilms are multimicrobial complex structures that are resistant to antibiotics and stressful environments. The biofilm stage may provide various advantages to the bacteria during bacterial infections in human beings. The extracellular polymeric substances hold the bacterial community colonized in the biofilm. The bacteria within the biofilm are more resistant to antibiotics, whereas the planktonic bacteria are susceptible to them. Quorum sensing regulated biofilm formation, which can be manipulated to eradicate devastating effects caused by biofilms. The occurrence of biofilm on the clinical devices leads to the malfunction of the implants and complicates the patients’ health conditions. Biofilms also cause non-device-associated health problems. The major anti-biofilm strategies are the utilization of enzymatic activity and hindrance of quorum sensing. The auto-inducers, which play a major role in quorum sensing, are mimicked by inhibitors. This prevents the binding of auto-inducers to the receptors, eventually leading to blockage of biofilm formation.

Short conclusion

The significant background knowledge regarding the biofilm, its formation, clinical aspects and inhibition strategies has been highlighted in this review. This information dissipated anticipates new applications of plant compounds as an alternative to antibiotics, since they may act as anti-quorum sensing molecules. For instance, inhibitory compounds like Linalool and eugenol from the essential oil of different plants displayed antibiofilm activity against biofilms formed by Streptococcus pyogenes and Porphyromonas gingivalis, respectively. Further research is required to exploit the inhibitory properties of the various other bioactive compounds present in plant extract, and thereby, we can protect human beings from several device and non-device-related infections caused by biofilms such as catheter-related bloodstream infections, tuberculosis, cystic fibrosis, chronic obstructive pulmonary diseases, dental caries and periodontitis.

Background

Biofilms are defined as a well-organized, multimicrobial three-dimensional community composed of a group of microorganisms encased within self-produced polymeric matrix, adhered to each other on biotic or abiotic surfaces. Initially, a small group of cells or a single cell divides and differentiates into a so-called complex structure [1]. The bacterial population within the biofilm can be homogenous or heterogeneous. Even though biofilms are frequently perceived as potentially harmful for the clinical and various other industrial domains, many biofilms are beneficial, and there are numerous reports about the beneficial use of these biofilms [2]. Beneficial biofilms have a variety of uses in agricultural, medical, environmental, food and other fields, including biofertilizer, antibacterial, antimicrobial agents, filtration, corrosion prevention, wastewater treatment biofouling, microbial fuel cells, bioremediation and food fermentation [2,3,4]. In cheese manufacturing, raw milk and Tina biofilm are the sources of the fermentative microflora which includes bacteria such as Lactobacillus lactis, Streptococcus thermophiles, Enterococcus faecium and Lactobacillus delbrueckii [5]. The primary bacteria that promote plant growth are Bacillus, Rhizobium, Acinetobacter, Enterobacter, Alcaligenes, Azospirillum, Burkholderia, Arthrobacter, Erwinia, Azotobacter, Flavobacterium, Serratia and Beijerinckia [6]. For a very long time, biofilms have been used successfully in the production of vinegar. Vinegar was produced on free-floating wood chips by Acetobacter or Gluconobacter with a higher output, which promotes the proliferation of vinegar bacteria [7].

Bioremediation is a method of pollution control in which various contaminants are destroyed or changed into less harmful forms. In bioremediation, microorganisms are used to break down environmental pollutants like plastic wastes, toxic heavy metals, synthetic dyes and other toxic compounds. Protection of the environment and human health depends on the bioremediation process [8]. Synthetic wastewater containing 2,4-dichlorophenol was degraded by P. putida, supplemented to the biofilm reactor with activated microbial sludge culture [9]. Alkanes, pesticides, polyaromatic substances and hydrocarbons can be broken down by aerobic microorganisms like Pseudomonas, Rhodococcus, Alcaligenes, Mycobacterium and Sphingomonas, and they use these pollutants to meet their needs for carbon sources and energy. In river sediments, anaerobic bacteria are used to break down polychlorinated biphenyls and dechlorinate the solvents trichloroethylene and chloroform [8]. The emergence and endurance of biofilms in the human body lead to the occurrence of above 75% of microbial infections, by the National Institutes of Health. Some bacterial biofilms have both functional and defensive roles, such as the gut microbiota [10]. Homeostasis has been maintained by interactions between the beneficial bacteria inside the intestine and the intestinal epithelium [11]. Dysbiosis is a state of imbalance in the microbiota, which is also related to various diseases [12].

Biofilms formed by bacteria play various roles in industries and are widely dispersed. In the dairy industry, equipment impairment and food spoilage caused by biofilms lead to economic loss for the industry as well as cause major hygienic problems (Fig. 1) [13]. Heat transfer efficiencies of the pipelines and heat exchangers can be reduced if a suitably thick layer of biofilms is formed; whereas corrosion in pipelines and tanks is caused by the biological and chemical reactions catalysed by the microbes present in the biofilms [14, 15]. Several bacterial populations producing biofilms display resistance against antibiotics [16]. The structure of the biofilms has been modulated to enable the bacteria to survive in different stressful environments [17]. The survival of single-celled organisms in adverse conditions has been facilitated by “group behaviour”, which is due to the biofilm that safeguards and promotes these organisms to lead a multicellular lifestyle [18]. On liquid surfaces, the biofilms are found to be seen as a floating mat and the biofilms are also found in submerged conditions [19]. Since the biofilms have diverse structural elements, both undifferentiated flat biofilms, as well as mushroom-like tall biofilms with distinct water channels, have been visualized, according to the type of bacteria responsible for their formation [20, 21].

Fig. 1
figure 1

Scanning electronic microscopy image of stainless steel of 3-day biofilms formed by Pseudomonas fluorescens. ©Carrascosa C et al. [22]

EPS are extracellular polymeric substances on which the bacterial communities are lodged. Polysaccharides are the main biomolecules present in EPS and it also consists of lipids, proteins and nucleic acids [23]. The bacterial community prevailing in the biofilm are held together by the scaffold formed by lipids, lipopolysaccharides and glycopeptides, which are polymers. The formation and stabilization of biofilms require cell-to-surface interactions and also interactions between the cells. This is mediated by bacterial polysaccharides [24]. It was discovered that viscoelastic behaviour, which helps to resist mechanical stress, is displayed by the biofilms and it was also revealed that they are hydrogels [25]. The water and nutrients are trapped in the matrix of EPS, which is used by the bacteria for its existence. Inside the matrix of EPS, the water is efficiently captured by its hydrogen bonding with the hydrophilic polysaccharides within the EPS [18]. Conceptual framework of the functions of microbial extracellular polymeric substances (EPS) in soil is depicted in Fig. 2

Fig. 2
figure 2

Conceptual framework of the functions of microbial extracellular polymeric substances (EPS) in soil. ©Costa OYA et al. [26]

Biofilms aid the bacteria to thrive in the environment by increasing their attachment to different surfaces; protecting them from desiccation, antibiotics, predation, immune attack and starvation. All these properties prevailing in the biofilms provide bacterial fitness to the concerned bacteria [27, 28]. Biofilm formation is regulated by quorum sensing; in addition to this quorum sensing also regulates spore formation and the production of secondary metabolites [29, 30]. Bacterial cell-to-cell communication is synchronously called quorum sensing, in which they communicate through auto-inducers (signalling molecules) in a density-dependent manner; it aids the coordination of the bacterial colony to exist as a sole component [31,32,33].

Main text

Types of biofilms

Based on the type of bacteria present in the biofilm, they are two types: Gram-positive biofilm and Gram-negative biofilm. On the cell walls of Gram-positive bacteria, teichoic acid (TA) is found, whereas in Gram-negative bacteria there is the presence of lipopolysaccharides (LPS). These components supply charges to the bacterial outer surface, which may also be responsible for the adherence to the surfaces and also in biofilm formation [34, 35]. The adhesion of the Gram-negative bacteria to the surface and their progression to biofilm formation is determined by the length of LPS and its dissimilarity in various parts of the bacteria [35]. Colonization of bacteria is influenced by teichoic acid in the case of Gram-positive bacteria and the case of Gram-negative bacteria, it is lipopolysaccharide that plays the role [36]. Cell wall teichoic acid present in Staphylococcus aureus plays a role in bacterial colonization on natural surfaces like nasal tissues and also on heart valve prostheses or catheters, which are artificial surfaces [34].

Cell wall surfaces appear to be a notable and precise target for anti-biofilm approaches since the surface of the cell wall governs the surface properties of two types of bacteria [37]. Both the Gram-type bacteria have flagella that helped them to move and are attached to the surface through polysaccharide secretion [38]. The Gram-positive flagellated bacteria (Bacillus subtilis) show attachment to surfaces through the exopolysaccharide formation, and its flagellar expressions are downregulated [39, 40], whereas its non-flagellated type (S. aureus) show Brownian motion since it has no organ for motility to reach the target surfaces [37]. The chemical nature of the quorum-sensing molecules varies by the production of AIP (autoinducing peptides) in Gram-positive bacteria and AHL (acyl-homoserine lactones) in Gram-negative bacteria [33]. Autoinducers-2 (AI-2) are non-species-specific and affect both Gram-positive and Gram-negative bacteria [41].

Mechanism of biofilm formation

There are four prominent phases in the sequential process of biofilm formation: (i) migration of cells and its adherence to the surface, (ii) micro-colony formation and EPS (extracellular polymeric substance) exudation from cells; irreversible attachment resulting in cell proliferation and formation of matrix, (iii) maturation stage, (iv) cell detachment [42]. The different stages of biofilms are represented in Fig. 3. Each of these steps is described below.

Fig. 3
figure 3

Mechanism of biofilm formation

Migration and adherence

In S. aureus, the initiation of biofilm happens when the floating cells are adhered to by the hydrophilic or hydrophobic interaction between the available surface and cell surface [43, 44]. The interaction of bacteria on the surface is strengthened by bacterial appendages like flagella, pili and fimbriae. This strengthening is due to the hydrophobic force on the surface, which causes the reduction of the repulsive force between the bacterial cell surface and the living or non-living surface [45]. Other than these forces, certain proteins also play a major role in mediating the binding between them. Heilmann and co-workers [46] first described such specific proteins, autolysins, which possess dual roles: adhesive and enzymatic function [47]. To bind to biotic surfaces, the cells produce certain CWA (Cell Wall-anchored) proteins. Among these MSCRAMMs (Microbial surface components recognizing adhesive matrix molecules) are the well-distinguished group [48]. A small amount of exopolymeric material is produced in the initial stage and the bacterial cells are encased in it [1].

Micro-colony formation and EPS

The bacteria attached to the surface proliferate and form micro-colonies. Different types of micro-communities are present in a biofilm. Metabolic product distribution, exchange of substrates and excretion are facilitated by coordination among the micro-colonies [49]. Constituents present in a bacterial matrix vary depending on the environmental conditions, different bacterial species and their strain types [50, 51]. The extracellular polymeric substance released by the cells contains polysaccharides, nucleic acids (eDNA), lipids and proteins [52]. This complex mixture is not only involved in structural support but also has a role in fetching nutrients and providing protection against antimicrobial agents and immune responses [53].

Maturation stage

Due to a change in microbial cell density, certain signalling molecules called auto-inducers are produced by these microbial cells. These auto-inducers play a vital role in quorum sensing (QS), the bacterial cell’s capability to sense the cell density through cell-to-cell communication. When a minimum threshold is attained, these signalling molecules induce alteration of gene expression [54]. The binding of the auto-inducer to the receptor causes the target genes to either be activated or repressed. Owing to this modification of the quorum sensing, the whole bacterial community in the biofilm benefits by exhibiting a combined response. This unified nature of the response is obtained by maintaining an ideal size of biofilm and synchronizing virulence phenotypes [55]. The integrated response of the bacteria enables it to act similarly to a multicellular organism, so it has the adaptation to live in a changing environment. In addition to managing bacterial population density, QS also assists the transfer of beneficial mutations among the colony, which provides tolerance to antibiotics and improves nutrient accessibility [56]. During the maturation stage, due to the action of auto-inducers, aggregation occurs between the bacterial cells by the secretion of EPS. A three-dimensional architecture is obtained by the biofilm and EPS is a prominent material responsible for this community. Voids are produced interstitially in the matrix and these structures act as channels which contain water. Nutrients are distributed and waste products are removed from the micro-colonies through this channel that also functions as a circulatory system [57].

Cell detachment

After completing the maturation, the cells in the biofilm produce certain chemicals and EPS lysing enzymes that degrade the matrix and help in the dispersion of the biofilm [18]. For this process of detachment, the cell communities within the biofilm use different degrading enzymes, for example, alginate lyase produced by Pseudomonas aeruginosa and P. fluorescens, hyaluronidase by Streptococcus equi and N-acetyl-heparosan lyase by Escherichia coli [58, 59]. Bacterial movement requires flagella. Therefore, protein expression of genes for flagella formation is upregulated and this allows the free locomotion of the organism, so that sessile cells are converted to free-floating motile cells [60, 61]. Finally, they can either form a new biofilm in a new area or they can get attached to the same region and recolonize [62].

Molecular mechanism

The decision of bacteria to either live as a planktonic form or to produce matrix and form biofilm is regulated by c-di-GMP signalling [63, 64]. The lower cellular level of c-di-GMP results in down regulation of matrix constituent production, which leads to the detachment of bacteria from the surface. Higher levels of c-di-GMP promotes matrix component production and induces biofilm formation [65, 66]. In some Gram-positive bacteria and in all Gram-negative bacteria, the major regulator of biofilm formation is c-di-GMP [64]. The bacteria produces certain enzymes such as DGCs (Diguanylate cyclase enzymes) and specific PDEs (Phosphodiesterase enzymes) which regulates the specificity of this signalling process with its physical interactions with c-di-GMP effectors. In bacteria, DGCs contributes to c-di-GMP formation while PDEs aids the degradation of c-di-GMP [67]. In accordance to the environmental conditions, bacteria can live in any of the two forms (sessile or planktonic) by regulation mediated through the enzyme’s regulatory domains in addition to its catalytic domains [68]. Both these enzymes, DGCs and PDEs have Gly-Gly-Asp-Glu-Phe (GGDEF) and Glu-Ala-Leu (EAL) / His-Asp-Gly-Tyr-Pro (HD-GYP) catalytic domains, respectively, which remains conserved [64]. Quorum sensing systems of Gram-negative bacteria use AHL (Acyl homoserine lactone) signal molecules produced by one or more AHL synthases. These signalling molecules can travel through the bacterial membranes and when they reach a threshold level of concentration, trigger one or more transcriptional factors and induce transcription of selective target genes. The correlation of signal molecule concentration to the bacterial population density enables the control of gene expression in a density-dependent manner [68, 69]. Quinolones are also employed as signalling molecules in both Gram-positive and Gram-negative bacteria. Luxl and LuxR homologue genes synthesizes HSL (Homoserine lactones), an auto-inducer which is also produced by Gram-negative bacteria. A transcriptional activator that senses the HSL is encoded by these homologous genes [70]. Some of the quorum sensing modulators are represented in Fig. 4.

Fig. 4
figure 4

Quorum sensing modulators

Clinical aspects

Although there have been advancements in the clinical field, biofilm infections caused by bacteria are still considered to be a major threat to human health. Lam, Hoiby and his teammates initially pointed out the fact that the recurrent infections are correlated with the biofilm formation, principally by Pseudomonas aeruginosa in cystic fibrosis patients [71, 72]. As time passed, the major source or cause for the tissue associated infections are accepted to be biofilms [73]. Even though there is a possibility of biofilm infections in humans due to hospital environment or previous existence, most of these tissue related biofilm illness have been developed in patients as a consequence of impaired immune system and chronic ailments like cancer, diabetes, cardiovascular disease, breakage of skin barrier [74]. The medical devices, most commonly pacemakers, peripheral vascular catheter that are used in the human body provides a surface for the bacteria to attach and form biofilms which eventually leads to infections; more over urinary infections, central line bloodstream infections, pacemaker infections, ventilator pneumonia are the other common device associated infections caused by biofilms [75, 76].

Even though the microbial antigens trigger the antibody production, there is no effective killing of bacteria present in the biofilm; the immune response by the antibody may also damage the tissues in its vicinity [77]. Antibiotic treatment do not affect the biofilm; it only affects the planktonic cells released from the biofilms [75]. Removal of the biofilm from the living tissue and the removal of the biofilm affected implants are the main ways to eradicate or avoid periodic infections [78]. Biofilms are responsible for various clinical infections such as dental caries, otitis media, periodontitis and osteomyelitis [60] and pulmonary infections seen in cystic fibrosis patients. Biofilms also causes health care associated infections, majorly including bacteremia, lower respiratory tract, surgical site and urinary tract infections which are developed due to pathogens that are opportunistic that mainly occur when a medical device has been inserted [10]. Catheters are much needed for the administration of drugs, blood products, nutrition and fluids into the veins; to filter the waste and water from the blood by haemodialysis; and to track the flow of blood within the organs and tissues of the body. The use of catheters paves way to a major health concern as it allows the growth of microbes on their surfaces, which eventually leads to infections causing serious health-related problems collectively named as CRBSIs (Catheter-related bloodstream infections). These infections were roughly calculated to amount to a total of 250,000 occurrences each year in the USA that are ruinous, involving a significant rate of disease and death in a population, in addition to an increase in health maintenance costs [79, 80].

The bacteria and microbes colonizing inside the catheter has the ability to avoid the host’s immune system and also the effect of antibiotics. Finally, the bacteria enters the blood stream by separating themselves from the biofilm, thus leading to metastatic infections and CRBSIs. Several measures are taken to reduce the rate of CRBSIs; among them, a major preventive measure is the development of locked catheter lumens using solutions with antimicrobial properties and using antimicrobial agent-coated catheter surfaces [10]. Biofilms also have its existence on tumour cells say in colorectal cancer. They may also be considered as the secondary cause for the tumour formation, since the initiation of tumour itself is due to the impairment of the mucus barrier [81] that may provide the conditions for the formation of biofilm. The tissue structure in the confined region is damaged because of the local immune response that occurs due to the presence of tumours. So the bacteria can effortlessly acquire its niche. Mucus production in tumour cells increases when there is a coordinated action between the bacterial biofilm and inflammation. Thus, the mucus released by these cells serves as a constituent for the formation of potent biofilms [82].

Bacterial biofilm infections involve both device-associated and non-device-associated infections. For example, pacemakers, mechanical heart valves, urinary catheters, peritoneal dialysis catheters, voice prostheses, central venous catheters, prosthetic joints and contact lenses are the examples of major medical devices placed inside the body that formed biofilms on the surface or inside the device [83]. There are two types of contact lenses: soft and hard, on which microbes can attach. Staphylococcus aureus, S. epidermidis, E. coli, P. aeruginosa, species of Serratia, Candida and Proteus, etc., are the microorganisms that adhere to the contact lenses. According to the nature of the substrate, water content, bacterial type, electrolyte concentration and polymer composition, the strength of adherence varies. The most frequent reason for lens contamination is biofilm formation in the contact lens store cases [84]. In consonance with the nature of the fluid delivered by means of a central venous catheter, the multiplication of microbes may be influenced; for instance, the effect varies in both types of bacteria; growth in the fluids are sustainable in some Gram-negative aquatic species like P. aeruginosa, Klebsiella species and Enterobacter species, whereas in some cases microbes do not show proper growth in intravenous fluids; these include Gram positive bacteria, like S. aureus and S. epidermidis [85, 86].

Bacteria such as S. epidermidis, S. aureus, Streptococcus species, Gram-negative Bacillus, Candida and Enterococcus species are attached to the mechanical heart valves and the tissues present in the surroundings and develops biofilm on them. This uncomfortable state is known as prosthetic valve endocarditis [87]. Pseudomonas aeruginosa, Klebsiella pneumoniae, E. coli, S. epidermidis, E. faecalis, Proteus mirabilis and other Gram-negative bacteria generally establish biofilms on urinary catheters [88]. This severely affects the health of the public and also affects instrumental and surgical procedures. Non-device-associated diseases produced by biofilms comprise persistent respiratory infections found in patients with cystic fibrosis, tuberculosis, chronic obstructive pulmonary diseases, chronic sinusitis, chronic wound infections, endocarditis, dental caries, chronic otitis media, biliary tract infections, osteomyelitis, bacterial prostatitis and periodontitis. Bacteria within the biofilm gained antibiotic resistance when there is only minimal regular exposure of antibiotics to the soft tissues inside the lungs or intestine. This minimal exposure may also affect the phenotypic and genetic variability in biofilm, bacterial physiology and also the potential of antibiotics to act as signalling molecules [89].

Non-device-associated infections also cause health issues. These infections occur in the oral gums, damage the bone where the teeth are held and in addition, affect the soft tissues. Poor oral hygiene causes such damage and tooth loss may also occur [90]. This infection is known as periodontitis in which the causative organisms are Porphyromonas gingivalis and Fusobacterium nucleatum, which can form biofilms in the oral cavity on the mucosal surfaces [91]. The entry of the bacteria into the bloodstream sometimes helps the bacteria enter the bones and infect the metaphysis of the bone, leading to osteomyelitis. Then WBCs are recruited to the site and they secrete enzymes to lyse the pathogens and to phagocytose them. Sometimes, these lysing enzymes may cause damage or lysis to the bone, forming pus and circulate it through the bone blood vessels. This causes a block in the normal blood flow, leading to the damage of tissues and loss of function in the affected area [92, 93].

One of the most important biofilms in the human body is dental plaque. On the surface of the teeth, diverse microbial communities build up. These plaques are contained in a polymeric matrix that is bacterial and salivary in origin [94]. The dental biofilms are primarily constituted by organic and inorganic materials on which micro-organisms exist. Dental plaques contain about 20% carbohydrates by dry weight, of which 2–10% constitutes homopolymers of glucose (glucans). The enzyme glucosyltransferases, extracellularly converts sucrose into glucans [95, 96]. Fructans (fructose polymers) are produced by fructosyltransferases from sucrose [97]. At the initial stage of plaque formation, Neisseria and Streptococci were the pioneer species. Most predominant among them were S. sanguis, S. oralis and S. mitis [94]. After the plaque was transformed into a mature community, the fraction of pioneer species steadily reduced while that of anaerobic bacteria including Fusobacterium, Veillonella, and Actinomyces grew. These anaerobic bacteria are established in the deeper layers of the plaque [98].

The bacteria inside the biofilms are in close proximity; therefore, the metabolic product (lactate) of the primary feeder (Streptococci) is utilized as a source of nutrition by the secondary feeder (Veillonella). This interaction can exert a controlling influence on the enamel demineralization by strong bacterial acids. Since lactate is converted to weak acids like acetic and propionic acids by Veillonella spp., caries causing ability of other plaque bacteria are reduced. Some oral bacteria produce inhibitory factors that are antagonistic in nature, among them bacteriocins are the most common antagonistic compound. Bacteriocins are bacterially derived antimicrobial peptides with high molecular weight. Examples of bacteriocins produced by oral streptococci are sanguicin produced by S. sanguis and mutacin by S. mutans. They are also produced by A. actinomycetemcomitans, black-pigmented anaerobes and C. matruchotii. Despite the fact that bacteriocins typically have a narrow range of action, several of the streptococcal bacteriocins have a broad spectrum of activity and can inhibit organisms from a variety of gram-positive genera, including Actinomyces spp. Similar to this, a bacteriocin from S. sanguis proved effective against both Gram-positive and Gram-negative bacteria, including species of Capnocytophaga and Prevotella. [94].

There is also another category of biofilm disease which is biofilm-related device malfunctioning. When a medical device is implanted, there is inflammatory and wound healing responses in the body since the device incorporated is a foreign body. There are several steps in a foreign body reaction [99] in which the surface properties of the biomaterial play a significant role in modulating these reactions. These modulations happen for two to four weeks once the medical device is implanted; but the foreign body reactions persists in the tissues and biomaterial contact regions until the implanted device is present inside the body [100]. The failure or malfunction of the clinical implant paves way to the appearance of mild symptoms like slight contracture or functional disability of soft tissue along with negative inflammatory markers and light pain [101, 102]. The malfunction of the biofilm associated medical device brings about disastrous clinical issues that includes physical damage and chemical degradation of the pacemaker leads, biliary tube blockage, malfunction of cerebrospinal fluid shunt, breast implant capsular contracture, crystalline encrustations on urinary stents, prosthetic joint failure and intravascular catheter malfunction. Finally, all these conditions lead to increase in morbidity and mortality, removal of implants and added hospital charges. Colonization of C. tropicalis and C. albicans on the transdermal endoscopic gastronomy feeding tubes give rise to polyurethane degeneration [103].

Inhibition of biofilm

Plant extract possess antibiofilm activity against biofilms produced by different microorganisms. The rhizome extract of Hedychium larsenii contained a compound Linalool in its essential oil that exhibited potential antibiofilm activity against Streptococcus pyogenes [104]. Eugenol obtained from the essential oil of Syzygium aromaticum (L.) Merr. & L. M. Perry (clove) leaf, suppressed the biofilm formation of an oral anaerobe Porphyromonas gingivalis [105]. The essential oil extracted from the leaves and flowers of Plumeria alba displayed good potential to inhibit the formation of Pseudomonas aeruginosa biofilms [106]. Most of the biofilm inhibitors are isolated from natural sources; some synthetic compounds, enzymes and chelating agents also possess anti-biofilm activity. Usnic acid, a secondary metabolite produced from lichen; synthetic halogenated furanones, bacteriophage-encoded endolysin (PlyC) and enzymes like Deoxyribonuclease I, glycoside hydrolase (dispersin B) are examples of other biofilm inhibitors [107,108,109,110]. Certain promising methods are developed from the existing hindrance mechanism that prevented biofilm formation, other than the common antibiotic therapy [111]. The two main novel strategies against the biofilm are the blockage of quorum sensing and enzyme-mediated biofilm inhibition.

By blockage of Quorum sensing

Quorum sensing (QS) has been considered as a significant mechanism in the synthesis of biofilms; hence, its blockage is one of the major strategies to inhibit the biofilm. The QS inhibitors prevent the communication among the bacteria, and this makes the bacteria sensitive to the antibiotic response and host immune system [112, 113]. Therefore, in order to control biofilm-associated infections, QS is considered as an effective target. Quorum quenching is the repression of QS sensing; in this mechanism, the communication signals are blocked by structural modification or by competitive inhibition, instead of degrading them [114, 115]. The antagonist of QS bound with signal molecules or with the receptor on which the signal molecules are usually bound; in either way, the receptor-signal molecule interaction has been disabled [116]. There are three main strategies used to block quorum sensing in Gram-negative bacteria: AHL biosynthesis blockage, AHL degradation or inactivation and signal receptor interference [117]. Various reactions take place sequentially during the AHL biosynthesis. To produce the homoserine lactone ring in the AHL, SAM (S-adenosyl methionine) is utilized. The acyl side chain in AHL is produced from an acyl carrier protein (ACP) precursor molecule that is charged sufficiently [118,119,120]. According to Zano et al. [121] and Masevicius and colleagues [122], for methylation processes, S-adenosyl-L-methionine (AdoMet) is produced by various Gram-negative bacteria as an initial methyl donor [117]. Two types of signal molecules involved in QS are synthesized from AdoMet and hence inhibition of AdoMet can cause the prevention of biofilm formation as reported by Zano et al. [121] [117]. S-adenosylhomocysteine and S-adenosylcysteine, analogous to SAM, are effective inhibitors that hindered the biosynthesis of AHL [118]. The AHL molecule biosynthesis of P. aeruginosa is shown to be inhibited by antibiotics like erythromycin and azithromycin when delivered at sub inhibitory concentrations. This suppression restricts the virulence factor and inhibits the biofilm formation [123,124,125].

AHL degradation or inactivation is a promising tactic to eliminate bacterial infections associated with biofilms. By enzyme activity, the AHL molecules are also degraded, thus avoiding the accumulation of AHL. The homoserine lactone ring of AHL is exposed without any disturbances to the rest of the molecule, by breaking the leftward bond of the double bonded oxygen with AHL lactonases and the bond present on the rightward of the double bonded oxygen is broken by enzyme decarboxylases. AiiA 240B1 produced by Bacillus spp. 24B1 is the first protein molecule identified to have the capability to hydrolyze the lactone ring of AHL molecule [126]. In the AHL molecule, the AHL acylase hydrolyzes the amide bond that is between the acyl side chain and homoserine lactone. This hydrolysis reaction in turn releases free fatty acid and homoserine lactone [127]. The carbon atoms in the acyl chain of this signal molecule are oxidized by utilizing AHL oxidase [128]. AHL signal molecules has not been broken by AHL oxidoreductases but has undergone alteration by which the binding efficiency of the signal molecule with the receptor proteins was modified [129].

Another strategy to prevent quorum sensing is the interruption of receptor molecules by analogous compounds. The physiological behaviours, mainly biofilm activity, virulence and antibacterial tolerance, are reduced in a bacterial community when the signal recognition is not happening. This is achieved when there is an interference in the binding of the AHL signal molecule with the receptor protein. The interruption is caused due to the binding of antagonist molecules with the receptors, making the receptor unavailable to the signal molecule for binding. In Staphylococcus epidermidis, an efficient QS inhibitor, furanone has been identified as an analogue molecule that successfully inhibits biofilm formation [130]. In some cases, the antimicrobial peptides also affect biofilm formation, besides the three strategies mentioned above. These peptides could hinder the QS system by either disturbing the transport of the signal molecules outside or within the cell, through which the cascade of signal transduction and the formation of biofilms are affected. CRISPRi is a recent technology applied in the latest research to minimize biofilm formation in Escherichia coli by blocking lux gene expression at the time of QS signalling [131]. Targeting c-di-GMP signalling systems is also considered as an efficient strategy to control biofilm [132].

By enzyme activity

Certain enzymes act as antibiofilm agents to remove the biofilm that had already formed. These include six main categories, such as oxidoreductases, transferases, synthetases, ligases, lyases and hydrolases. The cells from the bacterial biofilm are released from it when its disintegration happens due to the activity of saccharolytic enzymes yielded by some bacteria [133]. The enzymes block adhesive production and EPS formation, thereby inhibiting biofilm formation [134]. In biofilm formation, starch is a vital chemical component [135]; therefore, biofilm removal results due to polysaccharide degradation [133]. Biofilms of various bacteria are disrupted by some enzymes like protease, glycosidases and DNase, which are produced by the bacteria itself. These enzymes degrade the matrix, and therefore, the biofilms are dispersed. DNase has an inhibitory action on biofilms formed by both types of bacteria [136].

At the initial stage of biofilm formation for the existence of biofilm in a long run and to provide intactness, the extracellular DNA (e-DNA) play a significant role in the attachment of bacteria, matrix accumulation, stability, regulation and biofilm formation [137, 138]. According to Nijland and co-workers [139], the e-DNA in the EPS matrix of biofilm is dispersed by using DNase (NucB) [117]. In Staphylococcus species, biofilms have been reported to be inhibited by various proteases like trypsin, proteinase K, chymotrypsin, carboxypeptidase A and serratiopeptidase [140]. Alginate lyase produced from the marine bacterium source is considered an enzyme involved in biofilm disassembly [141,142,143,144]. Pseudoalteromonas is a marine bacterium from which the alginate lyase has been retrieved and it displays inhibitory action against biofilms formed by S. enterica, P. aeruginosa and E. coli [145, 146].

A potential antibiofilm activity has been found in amylase [147]. Alpha amylase aids the inactivation and eradication of the biofilm formation in S. aureus, as reported by Craigen and co-workers [148]. α amylase is considered as the most potent enzyme in polysaccharide inhibition from the investigation conducted by Divakaran et al. [149]. A combination of different enzymes is employed for biofilm eradication rather than using a single enzyme. The combined action of amylase, dextrin hydrolase and levan hydrolase has helped in biofilm removal [150]. Three enzymes like alpha-amylase, beta-glucanase and protease combined is found to be advantageous in industrial slime removal [151]. Various plant extracts are also reported to be effective against biofilm by having antivirulence, antiseptic and anti-QS properties and can damage the structure of mature biofilm and also prevent biofilm formation. Fresh Allium sativum extract displays competent activity against biofilms [152]. The different stages of biofilm formation and their inhibitors at each stage are represented in Fig. 5.

Fig. 5
figure 5

Different stages of biofilm formation and their inhibitors. Ghosh A et al. [153]. ©https://pubs.acs.org/doi/10.1021/acsomega.9b03695

Proposed mechanism

The enzymes like amylase break down the exopolysaccharide of bacteria that are attached to the surface as biofilms. The starch in the exopolysaccharide is broken down into sugars. Due to this, the bacteria in the biofilm are detached, and thus, further biofilm formation is inhibited. The exo-amylases and endo-amylases are the two groups formed by the classification of amylases on basis of the mode of action. End products formed are shorter when the substrates are hydrolyzed by exo-amylases and these enzymes act from the substrate’s non-reducing end [154], while oligosaccharides (end products) of different lengths are produced when the endo-amylases act unevenly on the glycosidic linkage present internal to the starch molecule [155]. Starch is a polysaccharide with amylose and amylopectin glucose polymers that are considered the main substrate on which α-1,4-glucan-4-glucanohydrolase efficiently acts. A disaccharide (maltose) and a monosaccharide (glucose) are produced when the α-1,4 and α-1,6-glycosidic bonds are hydrolysed with the help of α-amylase [133]. This enzyme needs Ca2+ metals for its stability since it is a metalloenzyme [156]. α-amylase can proceed with its action on the substrate from anywhere and can act faster, whereas β-amylase acts on α-1,4-glycosidic bond exclusively from the non-reducing end of starch. This is the cause for the higher inhibitory action of α-amylase compared to β-amylase [157]. Figure 6 depicts the proposed mechanism of biofilm inhibition.

Fig. 6
figure 6

Proposed mechanism of biofilm inhibition

Conclusions

Biofilm formation is an advantageous strategy employed by bacteria to protect themselves from antibiotics and to ensure their survival in stressful environmental conditions. Cell-to-cell and cell-to-surface adherences are assisted by the bacterial polysaccharide. The communication between the cells is regulated by quorum sensing, which utilizes auto-inducers as signalling molecules. The biofilms are pathogenic and cause several device and non-device-related clinical problems. To get rid of biofilms, either quorum sensing or c-di-GMP signalling is targeted. There are different enzymes which hinder the formation of extracellular polymeric substances and also disintegrate the matrix, which leads to the disassembly of biofilm. Plant extracts can also promote biofilm eradication since some of them have anti-QS compounds.

Availability of data and materials

The datasets used for the current study were collected from the journals, by using Google Scholar, Science Direct, PubMed, Springer link, JSTOR, etc. Conference papers, books were also used in this study.

Abbreviations

EPS:

Extracellular polymeric substances

TA:

Teichoic acid

LPS:

Lipopolysaccharides

AIP:

Autoinducing peptides

AHL:

Acyl-homoserine lactones

AI-2:

Autoinducers-2

CWA:

Cell wall-anchored

MSCRAMMs:

Microbial surface components recognizing adhesive matrix molecules

eDNA:

Extracellular DNA

QS:

Quorum sensing

DGCs:

Diguanylate cyclase enzyme

PDEs:

Phosphodiesterase enzymes

GGDEF:

Gly-Gly-Asp-Glu-Phe

EAL:

Glu-Ala-Leu

HD-GYP:

His-Asp-Gly-Tyr-Pro

HSL:

Homoserine lactones

CRBSIs:

Catheter-related bloodstream infections

SAM:

S-adenosyl methionine

ACP:

Acyl carrier protein

AdoMet:

S-adenosyl-L-methionine

References

  1. O’Toole GA, Kolter R (1998) Flagellar and twitching motility are necessary for Pseudomonas aeruginosa biofilm development. Mol Microbiol 30(2):295–304. https://doi.org/10.1046/j.1365-2958.1998.01062.x

    Article  CAS  PubMed  Google Scholar 

  2. Ünal Turhan E, Erginkaya Z, Korukluoğlu M, Konuray G (2019) Beneficial biofilm applications in food and agricultural industry. In: Malik A, Erginkaya Z, Erten H (eds) Health and safety aspects of food processing technologies. Springer, Cham, pp 445–469. https://doi.org/10.1007/978-3-030-24903-8_15.

  3. Qureshi N, Karcher P, Cotta M, Blaschek HP (2004) High-productivity continuous biofilm reactor for butanol production. Appl Biochem Biotechnol 114(1–3):713–721. https://doi.org/10.1385/ABAB:114:1-3:713

    Article  Google Scholar 

  4. Qureshi N (2009) Beneficial biofilms: wastewater and other industrial applications. In: Fratamico PM, Annous BA, Gunther NW (eds) Biofilms in the food and beverage industries. Woodhead Publishing Series in Food Science, Technology and Nutrition. Woodhead, Cambridge, pp 474–498.

  5. Licitra G, Ogier JC, Parayre S, Pediliggieri C, Carnemolla TM, Valentin H, Madec MN et al (2007) Variability of bacterial biofilms of the “tina” wood vats used in the ragusano cheese-making process. Appl Environ Microbiol 73(21):6980–6987. https://doi.org/10.1128/AEM.00835-07

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kasim WA, Gaafar RM, Ali RMA, Omar MN, Hewait HM (2016) Effect of biofilm forming plant growth promoting rhizobacteria on salinity tolerance in barley. Ann Agric Sci 61(2):217–227. https://doi.org/10.1016/j.aoas.2016.07.003

    Article  Google Scholar 

  7. Frazier WC, Westhoff DC (1988) Food microbiology, 4th edn. McGraw-Hill, New York, p 346

    Google Scholar 

  8. Vidali M (2001) Bioremediation. An overview. Pure Appl Chem 73(7):1163–1172. https://doi.org/10.1351/pac200173071163

    Article  CAS  Google Scholar 

  9. Kargi F, Eker S (2005) Removal of 2,4-dichlorophenol and toxicity from synthetic wastewater in a rotating perforated tube biofilm reactor. Process Biochem 40(6):2105–2111. https://doi.org/10.1016/j.procbio.2004.07.013

    Article  CAS  Google Scholar 

  10. Miquel S, Lagrafeuille R, Souweine B, Forestier C (2016) Anti-biofilm activity as a health issue. Front Microbiol 7:592. https://doi.org/10.3389/fmicb.2016.00592

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wrzosek L, Miquel S, Noordine ML, Bouet S, Chevalier-Curt MJ, Robert V, Philippe C et al (2013) Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol 11:61. https://doi.org/10.1186/1741-7007-11-61

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Martin R, Miquel S, Langella P, Bermúdez-Humarán LG (2014) The role of metagenomics in understanding the human microbiome in health and disease. Virulence 5(3):413–423. https://doi.org/10.4161/viru.27864

    Article  PubMed  PubMed Central  Google Scholar 

  13. Gram L, Bagge-Ravn D, Ng YY, Gymoese P, Vogel BF (2007) Influence of food soiling matrix on cleaning and disinfection efficiency on surface attached Listeria monocytogenes. Food Control 18(10):1165–1171. https://doi.org/10.1016/j.foodcont.2006.06.014

    Article  CAS  Google Scholar 

  14. Mittelman MW (1998) Structure and functional characteristics of bacterial biofilms in fluid processing operations. J Dairy Sci 81(10):2760–2764. https://doi.org/10.3168/jds.S0022-0302(98)75833-3

    Article  CAS  PubMed  Google Scholar 

  15. Vieira MJ, Melo LF, Pinheiro MM (1993) Biofilm formation: hydrodynamic effects on internal diffusion and structure. Biofouling 7(1):67–80. https://doi.org/10.1080/08927019309386244

    Article  CAS  Google Scholar 

  16. Brackman G, Cos P, Maes L, Nelis HJ, Coenye T (2011) Quorum sensing inhibitors increase the susceptibility of bacterial biofilms to antibiotics in vitro and in vivo. Antimicrob Agents Chemother 55(6):2655–2661. https://doi.org/10.1128/AAC.00045-11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Maric S, Vranes J (2007) Characteristics and significance of microbial biofilm formation. Period Biol 109:115–121

    Google Scholar 

  18. Kostakioti M, Hadjifrangiskou M, Hultgren SJ (2013) Bacterial biofilms: development, dispersal, and therapeutic strategies in the dawn of the postantibiotic era. Cold Spring Harb Perspect Med 3(4):a010306. https://doi.org/10.1101/cshperspect.a010306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lemon KP, Earl AM, Vlamakis HC, Aguilar C, Kolter R (2008) Biofilm Development with an Emphasis on Bacillus subtilis . In: Romeo T (eds) Bacterial biofilms. Current topics in microbiology and immunology, vol 322. Springer, Berlin. https://doi.org/10.1007/978-3-540-75418-3_1.

  20. Durham-Colleran MW, Verhoeven AB, van Hoek ML (2010) Francisella novicida forms in vitro biofilms mediated by an orphan response regulator. Microb Ecol 59(3):457–465. https://doi.org/10.1007/s00248-009-9586-9

    Article  PubMed  Google Scholar 

  21. Dean SN, Chung MC, van Hoek ML (2015) Burkholderia diffusible signal factor (BDSF) signals to Francisella novicida to disperse biofilm and increase siderophore production. Appl Environ Microbiol 81(20):7057–7066. https://doi.org/10.1128/AEM.02165-15

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Carrascosa C, Raheem D, Ramos F, Saraiva A, Raposo A (2021) Microbial biofilms in the food industry-a comprehensive review. Int J Environ Res Public Health 18(4):2014. https://doi.org/10.3390/ijerph18042014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cortes ME, Bonilla JC, Sinisterra RD (2011) Biofilm formation, control and novel strategies for eradication. In: Méndez-Vilas A (ed) Science against microbial pathogens: communicating current research and technological advances. Formatex Research Center, Badajoz, pp 896–905

    Google Scholar 

  24. Flemming HC, Wingender J (2010) The biofilm matrix. Nat Rev Microbiol 8:623–633. https://doi.org/10.1038/nrmicro2415

    Article  CAS  PubMed  Google Scholar 

  25. Stoodley P, Cargo R, Rupp CJ, Wilson S, Klapper I (2002) Biofilm material properties as related to shear-induced deformation and detachment phenomena. J Ind Microbiol Biotechnol 29:361–367. https://doi.org/10.1038/sj.jim.7000282

    Article  CAS  PubMed  Google Scholar 

  26. Costa OYA, Raaijmakers JM, Kuramae EE (2018) Microbial extracellular polymeric substances: ecological function and impact on soil aggregation. Front Microbiol 9:1636. https://doi.org/10.3389/fmicb.2018.01636

    Article  PubMed  PubMed Central  Google Scholar 

  27. Wong GCL, O’Toole GA (2011) All together now: integrating biofilm research across disciplines. MRS Bull 36(5):339–342. https://doi.org/10.1557/mrs.2011.64

    Article  PubMed  PubMed Central  Google Scholar 

  28. van Hoek ML (2013) Biofilms: an advancement in our understanding of Francisella species. Virulence 4(8):833–846. https://doi.org/10.4161/viru.27023

    Article  PubMed  PubMed Central  Google Scholar 

  29. Whitehead NA, Barnard AML, Slater H, Simpson NJL, Salmond GPC (2001) Quorum-sensing in Gram-negative bacteria. FEMS Microbiol Rev 25(4):365–404. https://doi.org/10.1111/j.1574-6976.2001.tb00583.x

    Article  CAS  PubMed  Google Scholar 

  30. Papenfort K, Bassler BL (2016) Quorum sensing signal-response systems in Gram-negative bacteria. Nat Rev Microbiol 14(9):576–588. https://doi.org/10.1038/nrmicro.2016.89

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fuqua WC, Winans SC, Greenberg EP (1994) Quorum sensing in bacteria: the Lux R-Lux I family of cell density-responsive transcriptional regulators. J Bacteriol 176(2):269–275. https://doi.org/10.1128/jb.176.2.269-275.1994

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Parker CT, Sperandio V (2009) Cell-to-cell signalling during pathogenesis. Cell Microbiol 11(3):363–369. https://doi.org/10.1111/j.1462-5822.2008.01272.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Whiteley M, Diggle SP, Greenberg EP (2017) Progress in and promise of bacterial quorum sensing research. Nature 551:313–320. https://doi.org/10.1038/nature24624

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Weidenmaier C, Kokai-Kun JF, Kristian SA, Chanturiya T, Kalbacher H, Gross M, Nicholson G et al (2004) Role of teichoic acids in Staphylococcus aureus nasal colonization, a major risk factor in nosocomial infections. Nat Med 10(3):243–245. https://doi.org/10.1038/nm991

    Article  CAS  PubMed  Google Scholar 

  35. Lee KJ, Lee MA, Hwang W, Park H, Lee KH (2016) Deacylated lipopolysaccharides inhibit biofilm formation by Gram-negative bacteria. Biofouling 32(7):711–723. https://doi.org/10.1080/08927014.2016.1193595

    Article  CAS  PubMed  Google Scholar 

  36. Bucher T, Oppenheimer-Shaanan Y, Savidor A, Bloom-Ackermann Z, Kolodkin-Gal I (2015) Disturbance of the bacterial cell wall specifically interferes with biofilm formation. Environ Microbiol Rep 7(6):990–1004. https://doi.org/10.1111/1758-2229.12346

    Article  CAS  PubMed  Google Scholar 

  37. Ruhal R, Kataria R (2021) Biofilm patterns in gram-positive and gram-negative bacteria. Microbiol Res 251:126829. https://doi.org/10.1016/j.micres.2021.126829

    Article  CAS  PubMed  Google Scholar 

  38. Zhao K, Tseng BS, Beckerman B, Jin F, Gibiansky ML, Harrison JJ, Luijten E et al (2013) Psl trails guide exploration and microcolony formation in Pseudomonas aeruginosa biofilms. Nature 497:388–391. https://doi.org/10.1038/nature12155

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Blair KM, Turner L, Winkelman JT, Berg HC, Kearns DB (2008) A molecular clutch disables flagella in the Bacillus subtilis biofilm. Science 320(5883):1636–1638. https://doi.org/10.1126/science.1157877

    Article  CAS  PubMed  Google Scholar 

  40. Guttenplan SB, Blair KM, Kearns DB (2010) The EpsE flagellar clutch is bifunctional and synergizes with EPS biosynthesis to promote Bacillus subtilis biofilm formation. PLoS Genet 6(12):e1001243. https://doi.org/10.1371/journal.pgen.1001243

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Miller MB, Bassler BL (2001) Quorum sensing in bacteria. Annu Rev Microbiol 55:165–199. https://doi.org/10.1146/annurev.micro.55.1.165

    Article  CAS  PubMed  Google Scholar 

  42. Sutherland IW (2001) The biofilm matrix—an immobilized but dynamic microbial environment. Trends Microbiol 9(5):222–227. https://doi.org/10.1016/S0966-842X(01)02012-1

    Article  CAS  PubMed  Google Scholar 

  43. Hamadi F, Latrache H, Mabrrouki M, Elghmari A, Outzourhit A, Ellouali M, Chtaini A (2005) Effect of pH on distribution and adhesion of Staphylococcus aureus to glass. J Adhes Sci Technol 19(1):73–85. https://doi.org/10.1163/1568561053066891

    Article  CAS  Google Scholar 

  44. Maikranz E, Spengler C, Thewes N, Thewes A, Nolle F, Jung P, Bischoff M et al (2020) Different binding mechanisms of Staphylococcus aureus to hydrophobic and hydrophilic surfaces. Nanoscale 12(37):19267–19275. https://doi.org/10.1039/D0NR03134H

    Article  CAS  PubMed  Google Scholar 

  45. Tribedi P, Sil AK (2014) Cell surface hydrophobicity: a key component in the degradation of polyethylene succinate by Pseudomonas sp. AKS2. J Appl Microbiol 116(2):295–303. https://doi.org/10.1111/jam.12375

    Article  CAS  PubMed  Google Scholar 

  46. Heilmann C, Hussain M, Peters G, Gotz F (1997) Evidence for autolysin-mediated primary attachment of Staphylococcus epidermidis to a polystyrene surface. Mol Microbiol 24(5):1013–1024. https://doi.org/10.1046/j.1365-2958.1997.4101774.x

    Article  CAS  PubMed  Google Scholar 

  47. Foster SJ (1995) Molecular characterization and functional analysis of the major autolysin of Staphylococcus aureus 8325/4. J Bacteriol 177(19):5723–5725. https://doi.org/10.1128/jb.177.19.5723-5725.1995

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schilcher K, Horswill AR (2020) Staphylococcal biofilm development: structure, regulation, and treatment strategies. Microbiol Mol Biol Rev 84(3):1–36. https://doi.org/10.1128/MMBR.00026-19

    Article  Google Scholar 

  49. Caldwell DE (1995) Cultivation and study of biofilm communities. In: Scott HML, Costerton JW (eds) Microbial biofilms. University Press, Cambridge, pp 64–79

    Chapter  Google Scholar 

  50. Stewart PS, Costerton JW (2001) Antibiotic resistance of bacteria in biofilms. Lancet 358(9276):135–138. https://doi.org/10.1016/S0140-6736(01)05321-1

    Article  CAS  PubMed  Google Scholar 

  51. Arciola CR, Campoccia D, Gamberini S, Donati ME, Pirini V, Visai L, Speziale P et al (2005) Antibiotic resistance in exopolysaccharide-forming Staphylococcus epidermidis clinical isolates from orthopaedic implant infections. Biomaterials 26(33):6530–6535. https://doi.org/10.1016/j.biomaterials.2005.04.031

    Article  CAS  PubMed  Google Scholar 

  52. Annous BA, Fratamico PM, Smith JL (2009) Quorum sensing in biofilms: why bacteria behave the way they do. J Food Sci. https://doi.org/10.1111/j.1750-3841.2008.01022.x

    Article  PubMed  Google Scholar 

  53. Flemming HC, Neu TR, Wozniak DJ (2007) The EPS matrix: the “house of biofilm cells.” J Bacteriol 189(22):7945–7947. https://doi.org/10.1128/JB.00858-07

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Davies DG, Parsek MR, Pearson JP, Iglewski BH, Costerton JW, Greenberg EP (1998) The involvement of cell-to-cell signals in the development of a bacterial biofilm. Science 280(5361):295–298. https://doi.org/10.1126/science.280.5361.295

    Article  CAS  PubMed  Google Scholar 

  55. Nadell CD, Xavier JB, Levin SA, Foster KR (2008) The evolution of quorum sensing in bacterial biofilms. PLoS Biol. https://doi.org/10.1371/journal.pbio.0060014

    Article  PubMed  PubMed Central  Google Scholar 

  56. Hannan S, Ready D, Jasni AS, Rogers M, Pratten J, Roberts AP (2010) Transfer of antibiotic resistance by transformation with eDNA within oral biofilms. Immunol Med Microbiol 59(3):345–349. https://doi.org/10.1111/j.1574-695X.2010.00661.x

    Article  CAS  Google Scholar 

  57. Parsek MR, Singh PK (2003) Bacterial biofilms: an emerging link to disease pathogenesis. Annu Rev Microbiol 57:677–701. https://doi.org/10.1146/annurev.micro.57.030502.090720

    Article  CAS  PubMed  Google Scholar 

  58. Schiller NL, Monday SR, Boyd C, Keen NT, Ohman DE (1993) Characterization of the Pseudomonas alginate lyase gene (algL): cloning, sequencing and expression in Escherichia coli. J Bacteriol 175(15):4780–4789. https://doi.org/10.1128/jb.175.15.4780-4789.1993

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Legoux R, Lelong P, Jourde C, Feuillerat C, Capdevielle J, Sure V, Ferran E et al (1996) N-acetyl-heparosan lyase of Escherichia coli K5: gene cloning and expression. J Bacteriol 178(24):7260–7264. https://doi.org/10.1128/jb.178.24.7260-7264.1996

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Costerton JW, Stewart PS, Greenberg EP (1999) Bacterial biofilms: a common cause of persistent infections. Science 284(5418):1318–1322. https://doi.org/10.1126/science.284.5418.1318

    Article  CAS  PubMed  Google Scholar 

  61. Otto M (2013) Staphylococcal infections: mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu Rev Med 64:175–188. https://doi.org/10.1146/annurev-med-042711-140023

    Article  CAS  PubMed  Google Scholar 

  62. Donlan RM (2002) Biofilms: microbial life on surfaces. Emerg Infect Dis 8(9):881–890

    Article  PubMed  PubMed Central  Google Scholar 

  63. Fazli M, Almblad H, Rybtke ML, Givskov M, Eberl L, Tolker-Nielsen T (2014) Regulation of biofilm formation in Pseudomonas and Burkholderia species. Environ Microbiol 16(7):1961–1981. https://doi.org/10.1111/1462-2920.12448

    Article  CAS  PubMed  Google Scholar 

  64. Jenal U, Reinders A, Lori C (2017) Cyclic di-GMP: second messenger extraordinaire. Nat Rev Microbiol 15(5):271–284. https://doi.org/10.1038/nrmicro.2016.190

    Article  CAS  PubMed  Google Scholar 

  65. Gjermansen M, Ragas P, Tolker-Nielsen T (2006) Proteins with GGDEF and EAL domains regulate Pseudomonas putida biofilm formation and dispersal. FEMS Microbiol Lett 265(2):215–224. https://doi.org/10.1111/j.1574-6968.2006.00493.x

    Article  CAS  PubMed  Google Scholar 

  66. Christensen LD, van Gennip M, Rybtke MT, Wu H, Chiang WC, Alhede M, Hoiby N et al (2013) Clearance of Pseudomonas aeruginosa foreign-body biofilm infections through reduction of the cyclic di-GMP Level in the bacteria. Infect Immun 81(8):2705–2713. https://doi.org/10.1128/IAI.00332-13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sarenko O, Klauck G, Wilke FM, Pfiffer V, Richter AM, Herbst S, Kaever V, Hengge R (2017) More than enzymes that make or break cyclic di-GMP-local signaling in the interactome of GGDEF/EAL domain proteins of Escherichia coli. MBio 8(5):1–18. https://doi.org/10.1128/mBio.01639-17

    Article  Google Scholar 

  68. Qvortrup K, Hultqvist LD, Nilsson M, Jakobsen TH, Jansen CU, Uhd J, Andersen JB et al (2019) Small molecule anti-biofilm agents developed on the basis of mechanistic understanding of biofilm formation. Front Chem 7:742. https://doi.org/10.3389/fchem.2019.00742

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Vadakkan K, Vijayanand S, Hemapriya J, Gunasekaran R (2019) Quorum sensing inimical activity of Tribulus terrestris against gram negative bacterial pathogens by signalling interference. 3 Biotech 9:163. https://doi.org/10.1007/s13205-019-1695-7

    Article  PubMed  PubMed Central  Google Scholar 

  70. Lahiri D, Dash S, Dutta R, Nag M (2019) Elucidating the effect of anti-biofilm activity of bioactive compounds extracted from plants. J Biosci 44(52):1–19. https://doi.org/10.1007/s12038-019-9868-4

    Article  Google Scholar 

  71. Lam J, Chan R, Lam K, Costerton JW (1980) Production of mucoid microcolonies by Pseudomonas aeruginosa within infected lungs in cystic fibrosis. Infect Immun 28(2):546–556. https://doi.org/10.1128/iai.28.2.546-556.1980

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Høiby N (1977) Pseudomonas aeruginosa infection in cystic fibrosis: Diagnostic and prognostic significance of Pseudomonas aeruginosa precipitins determined by means of crossed immunoelectrophoresis. A survey. Acta Pathol Microbiol Scand Sect C Immunol 85 Suppl(262):1–96

    Google Scholar 

  73. Lebeaux D, Chauhan A, Rendueles O, Beloin C (2013) From in vitro to in vivo models of bacterial biofilm-related infections. Pathogens 2(2):288–356. https://doi.org/10.3390/pathogens2020288

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sivaranjani M, Srinivasan R, Aravindraja C, Karutha Pandian S, Veera Ravi A (2018) Inhibitory effect of α-mangostin on Acinetobacter baumannii biofilms-an in vitro study. Biofouling 34(5):579–593. https://doi.org/10.1080/08927014.2018.1473387

    Article  CAS  PubMed  Google Scholar 

  75. Marrie TJ, Nelligan J, Costerton JW (1982) A scanning and transmission electron microscopic study of an infected endocardial pacemaker lead. Circulation 66(6):1339–1341. https://doi.org/10.1161/01.cir.66.6.1339

    Article  CAS  PubMed  Google Scholar 

  76. Kannappan A, Srinivasan R, Nivetha A, Annapoorani A, Pandian SK, Ravi AV (2019) Anti-virulence potential of 2-hydroxy-4-methoxybenzaldehyde against methicillin-resistant Staphylococcus aureus and its clinical isolates. Appl Microbiol Biotechnol 103(16):6747–6758. https://doi.org/10.1007/s00253-019-09941-6

    Article  CAS  PubMed  Google Scholar 

  77. Cochrane DMG, Brown MRW, Anwar H, Weller PH, Lam K, Costerton JW (1988) Antibody response to Pseudomonas aeruginosa surface protein antigens in a rat model of chronic lung infection. J Med Microbiol 27(4):255–261. https://doi.org/10.1099/00222615-27-4-255

    Article  CAS  PubMed  Google Scholar 

  78. Costerton JW, Lewandowski Z, Caldwell DE, Korber DR, Lappin-Scott HM (1995) Microbial biofilms. Annu Rev Microbial 49:711–745. https://doi.org/10.1146/annurev.mi.49.100195.003431

    Article  CAS  Google Scholar 

  79. Maki DG (1994) Infections caused by intravascular devices used in infusion therapy: pathogenesis, prevention, and management. In: Bisno AL, Waldvogel FA (eds) Infections associated with indwelling medical devices. ASM Press, Washington, DC, pp 155–212

    Google Scholar 

  80. Crnich CJ, Maki DG (2005) Infections caused by intravascular devices: epidemiology, pathogenesis, diagnosis, prevention, and treatment. In: APIC text of infection control and epidemiology, vol 1. Association for Professionals in Infection Control and Epidemiology, Inc, Washington, DC.

  81. Matsuo K, Ota H, Akamatsu T, Sugiyama A, Katsuyama T (1997) Histochemistry of the surface mucous gel layer of the human colon. Gut 40(6):782–789. https://doi.org/10.1136/gut.40.6.782

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Li S, Peppelenbosch MP (1872) Smits R (2019) Bacterial biofilms as a potential contributor to mucinous colorectal cancer formation. Biochim Biophys Acta Rev Cancer 1:74–79. https://doi.org/10.1016/j.bbcan.2019.05.009

    Article  CAS  Google Scholar 

  83. Donlan RM (2001) Biofilms and device-associated infections. Emerg Infect Dis 7(2):277–281

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Donlan RM, Costerton JW (2002) Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev 15(2):167–193. https://doi.org/10.1128/cmr.15.2.167-193.2002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Maki DG, Mermel LA (1998) Infections due to infusion therapy. In: Bennett JV, Brachman PS (eds) Hospital infections. Lippincott-Raven publishers, Philadelphia, PA, pp 689–724

    Google Scholar 

  86. Donlan RM, Murga R, Carson L (1999) Growing biofilms in intravenous fluids. In: Wimpenny JWT, Gilbert P, Walker J, Brading M, Bayston R (eds) Biofilms: the good, the bad, and the ugly. Bioline, Wales, pp 23–29

    Google Scholar 

  87. Braunwald E (1997) Valvular heart disease. In: Braunwald E (ed) Heart disease. W.B. Saunders Co, Philadelphia, pp 1007–1066

    Google Scholar 

  88. Stickler DJ (1996) Bacterial biofilms and the encrustation of urethral catheters. Biofouling 9(4):293–305. https://doi.org/10.1080/08927019609378311

    Article  Google Scholar 

  89. Andersson DI, Hughes D (2014) Microbiological effects of sublethal levels of antibiotics. Nat Rev Microbiol 12(7):465–478. https://doi.org/10.1038/nrmicro3270

    Article  CAS  PubMed  Google Scholar 

  90. Jamal M, Ahmad W, Andleeb S, Jalil F, Imran M, Nawaz MA, Hussain T et al (2018) Bacterial biofilm and associated infections. J Chin Med Assoc 81(1):7–11. https://doi.org/10.1016/j.jcma.2017.07.012

    Article  PubMed  Google Scholar 

  91. Lamont RJ, Jenkinson HF (1998) Life below the gum line: pathogenic mechanisms of Porphyromonas gingivalis. Microbiol Mol Biol Rev 62(4):1244–1263. https://doi.org/10.1128/MMBR.62.4.1244-1263.1998

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ziran BH (2007) Osteomyelitis. J Trauma 62(6 Suppl):S59-60. https://doi.org/10.1097/TA.0b013e318065abbd

    Article  PubMed  Google Scholar 

  93. Kumar V, Abbas AK, Fausto N, Mitchell RN (2007) Robbins basic pathology, 8th edn. Saunders Elsevier, Philadelphia, pp 810–811

    Google Scholar 

  94. Marsh P, Martin M (1992) Dental plaque. In: Oral microbiology. Aspects of microbiology, vol 1. Springer, Boston, MA, pp 98–132. https://doi.org/10.1007/978-1-4615-7556-6_5.

  95. Hotz P, Guggenheim B, Schmid R (1972) Carbohydrates in pooled dental plaque. Caries Res 6:103–121. https://doi.org/10.1159/000259783

    Article  CAS  PubMed  Google Scholar 

  96. Wood JM (1969) The state of hexose sugar in human dental plaque and its metabolism by the plaque bacteria. Arch Oral Biol 14(2):161–168. https://doi.org/10.1016/0003-9969(69)90060-0

    Article  CAS  PubMed  Google Scholar 

  97. van Hijum SA, Kralj S, Ozimek LK, Dijkhuizen L, van Geel-Schutten IG (2006) Structure-function relationships of glucansucrase and fructansucrase enzymes from lactic acid bacteria. Microbiol Mol Biol Rev 70(1):157–176. https://doi.org/10.1128/MMBR.70.1.157-176.2006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ritz HL (1967) Microbial population shifts in developing human dental plaque. Arch Oral Biol 12(12):1561–1568. https://doi.org/10.1016/0003-9969(67)90190-2

    Article  CAS  PubMed  Google Scholar 

  99. Adams WP Jr, Haydon MS, Raniere J Jr, Trott S, Marques M, Feliciano M, Robinson JB Jr et al (2006) A rabbit model for capsular contracture: development and clinical implications. Plast Reconstr Surg 117(4):1214–1219. https://doi.org/10.1097/01.prs.0000208306.79104.18

    Article  CAS  PubMed  Google Scholar 

  100. Glage S, Paret S, Winkel A, Stiesch M, Bleich A, Krauss JK, Schwabe K (2017) A new model for biofilm formation and inflammatory tissue reaction: intraoperative infection of a cranial implant with Staphylococcus aureus in rats. Acta Neurochir 159(9):1747–1756. https://doi.org/10.1007/s00701-017-3244-7

    Article  PubMed  Google Scholar 

  101. Del Pozo JL, Tran NV, Petty PM, Johnson CH, Walsh MF, Bite U, Clay RP et al (2009) Pilot study of association of bacteria on breast implants with capsular contracture. J Clin Microbiol 47(5):1333–1337. https://doi.org/10.1128/JCM.00096-09

    Article  PubMed  PubMed Central  Google Scholar 

  102. Romano CL, Romano D, Morelli I, Drago L (2016) The concept of biofilm-related implant malfunction and “low-grade infection”. In: Drago L (ed) A modern approach to biofilm-related orthopaedic implant infections. advances in experimental medicine and biology, vol 971. Springer, Cham, pp 1–13. https://doi.org/10.1007/5584_2016_158.

  103. Trevisani L, Sartori S, Rossi MR, Bovolenta R, Scoponi M, Gullini S, Abbasciano V (2005) Degradation of polyurethane gastrostomy devices: what is the role of fungal colonization? Dig Dis Sci 50(3):463–469. https://doi.org/10.1007/s10620-005-2459-2

    Article  PubMed  Google Scholar 

  104. Praseetha S, Sukumaran ST, Dan M, Augustus AR, Pandian SK, Sugathan S (2023) The anti-biofilm potential of Linalool, a major compound from Hedychium larsenii, against Streptococcus pyogenes and its toxicity assessment in Danio rerio. Antibiotics 12(3):545. https://doi.org/10.3390/antibiotics12030545

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Zhang Y, Wang Y, Zhu X, Cao P, Wei S, Lu Y (2017) Antibacterial and antibiofilm activities of eugenol from essential oil of Syzygium aromaticum (L.) Merr. & L. M. Perry (clove) leaf against periodontal pathogen Porphyromonas gingivalis. Microb Pathog 113:396–402. https://doi.org/10.1016/j.micpath.2017.10.054

    Article  CAS  PubMed  Google Scholar 

  106. Mamattah KMM, Adomako AK, Mensah CN, Borquaye LS (2023) Chemical characterization, antioxidant, antimicrobial, and antibiofilm activities of essential oils of Plumeria alba (Forget-Me-Not). Biochem Res Int 2023:1040478. https://doi.org/10.1155/2023/1040478

    Article  Google Scholar 

  107. Hentzer M, Riedel K, Rasmussen TB, Heydorn A, Anderson JB, Parsck MR, Rice SA et al (2002) Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm bacteria by a halogenated furanone compound. Microbiology 148(1):87–102. https://doi.org/10.1099/00221287-148-1-87

    Article  CAS  PubMed  Google Scholar 

  108. Francolini I, Norris P, Piozzi A, Donelli G, Stoodley P (2004) Usnic acid, a natural antimicrobial agent able to inhibit bacterial biofilm formation on polymer surfaces. Antimicrob Agents Chemother 48(11):4360–4365. https://doi.org/10.1128/AAC.48.11.4360-4365.2004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Donelli G, Francolini I, Romoli D, Guaglianone E, Piozzi A, Ragunath C, Kaplan JB (2007) Synergistic activity of dispersin B and cefamandole nafate in inhibition of staphylococcal biofilm growth on polyurethanes. Antimicrob Agents Chemother 51(8):2733–2740. https://doi.org/10.1128/AAC.01249-06

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Shen Y, Koller T, Kreikemeyer B, Nelson DC (2013) Rapid degradation of Streptococcus pyogenes biofilms by PlyC, a bacteriophage-encoded endolysin. J Antimicrob Chemother 68(8):1818–1824. https://doi.org/10.1093/jac/dkt104

    Article  CAS  PubMed  Google Scholar 

  111. Shunmugaperumal T (2010) Biofilm eradication and prevention: a pharmaceutical approach to medical device infections, 1st edn. Wiley, Hoboken

    Book  Google Scholar 

  112. Ravindran D, Ramanathan S, Arunachalam K, Jeyaraj GP, Shunmugiah KP, Arumugam VR (2018) Phytosynthesized silver nanoparticles as antiquorum sensing and antibiofilm agent against the nosocomial pathogen Serratia marcescens: an in vitro study. J Appl Microbiol 124(6):1425–1440. https://doi.org/10.1111/jam.13728

    Article  CAS  PubMed  Google Scholar 

  113. Li D, Ramanathan S, Wang G, Wu Y, Tang Q, Li G (2020) Acetylation of lysine 7 of AhyI affects the biological function in Aeromonas hydrophila. Microb Pathog 140:103952. https://doi.org/10.1016/j.micpath.2019.103952

    Article  CAS  PubMed  Google Scholar 

  114. Chan KG, Liu YC, Chang CY (2015) Inhibiting N-acyl-homoserine lactone synthesis and quenching Pseudomonas quinolone quorum sensing to attenuate virulence. Front Microbiol 6:1173. https://doi.org/10.3389/fmicb.2015.01173

    Article  PubMed  PubMed Central  Google Scholar 

  115. Vadakkan K, Vijayanand S, Hemapriya J, Gunasekaran R (2022) Quorum quenching attenuation of rhizobacteria Chromobacterium violaceum virulence by Desmodium gangeticum root extract. Rhizosphere 22:100490. https://doi.org/10.1016/j.rhisph.2022.100490

    Article  Google Scholar 

  116. Vadakkan K, Choudhury AA, Gunasekaran R, Hemapriya J, Vijayanand S (2018) Quorum sensing intervened bacterial signaling: pursuit of its cognizance and repression. J Genet Eng Biotechnol 16(2):239–252. https://doi.org/10.1016/j.jgeb.2018.07.001

    Article  PubMed  PubMed Central  Google Scholar 

  117. Srinivasan R, Santhakumari S, Poonguzhali P, Geetha M, Dyavaiah M, Xiangmin L (2021) Bacterial biofilm inhibition: a focused review on recent therapeutic strategies for combating the biofilm mediated infections. Front Microbiol 12:676458. https://doi.org/10.3389/fmicb.2021.676458

    Article  PubMed  PubMed Central  Google Scholar 

  118. Parsek MR, Val DL, Hanzelka BL, Cronan JE Jr, Greenberg EP (1999) Acyl homoserine-lactone quorum-sensing signal generation. Proc Natl Acad Sci 96(8):4360–4365. https://doi.org/10.1073/pnas.96.8.4360

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Hentzer M, Givskov M (2003) Pharmacological inhibition of quorum sensing for the treatment of chronic bacterial infections. J Clin Invest 112(9):1300–1307. https://doi.org/10.1172/JCI20074

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Li W, Yao Z, Sun L, Hu W, Cao J, Lin WX, Lin X (2016) Proteomics analysis reveals a potential antibiotic cocktail therapy strategy for Aeromonas hydrophila infection in biofilm. J Proteome Res 15(6):1810–1820. https://doi.org/10.1021/acs.proteome.5b01127

    Article  CAS  PubMed  Google Scholar 

  121. Zano SP, Bhansali P, Luniwal A, Viola RE (2013) Alternative substrates selective for S-adenosylmethionine synthetases from pathogenic bacteria. Arch Biochem Biophys 536(1):64–71. https://doi.org/10.1016/j.abb.2013.05.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Masevicius V, Nainytė M, Klimasauskas S (2016) Synthesis of S-Adenosyl-L-Methionine analogs with extended transferable groups for methyltransferase-directed labeling of DNA and RNA. Curr Protoc Nucleic Acid Chem 64:1361–13613. https://doi.org/10.1002/0471142700.nc0136s64

    Article  Google Scholar 

  123. Sofer D, Gilboa-Garber N, Belz A, Garber NC (1999) ‘Subinhibitory’ erythromycin represses production of Pseudomonas aeruginosa lectins, autoinducer and virulence factors. Chemotherapy 45(5):335–341. https://doi.org/10.1159/000007224

    Article  CAS  PubMed  Google Scholar 

  124. Tateda K, Comte R, Pechere JC, Kohler T, Yamaguchi K, Van Delden C (2001) Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrob Agents Chemother 45(6):1930–1933. https://doi.org/10.1128/aac.45.6.1930-1933.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Pechere JC (2001) Azithromycin reduces the production of virulence factors in Pseudomonas aeruginosa by inhibiting quorum sensing. Jpn J Antibiot 54(Suppl C):87–89

    PubMed  Google Scholar 

  126. Dong YH, Xu JL, Li XZ, Zhang LH (2000) AiiA, an enzyme that inactivates the acylhomoserine lactone quorum-sensing signal and attenuates the virulence of Erwinia carotovora. Proc Natl Acad Sci 97(7):3526–3531. https://doi.org/10.1073/pnas.97.7.3526

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Leadbetter JR, Greenberg EP (2000) Metabolism of acyl-homoserine lactone quorum-sensing signals by Variovorax paradoxus. J Bacteriol 182(24):6921–6926. https://doi.org/10.1128/jb.182.24.6921-6926.2000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Gao A, Mei GY, Liu S, Wang P, Tang Q, Liu YP, Wen H et al (2013) High-resolution structures of AidH complexes provide insights into a novel catalytic mechanism for N-acyl homoserine lactonase. Acta Crystallogr D Biol Crystallogr 69:82–91. https://doi.org/10.1107/s0907444912042369

    Article  CAS  PubMed  Google Scholar 

  129. Vogel J, Quax WJ (2019) Enzymatic quorum quenching in biofilms. In: Tommonaro G (ed) Quorum sensing: molecular mechanism and biotechnological application. Academic Press, pp 173–193. doi:https://doi.org/10.1016/b978-0-12-814905-8.00007-1.

  130. Hume EBH, Baveja J, Muir B, Schubert TL, Kumar N, Kjelleberg S, Griesser HJ et al (2004) The control of Staphylococcus epidermidis biofilm formation and in vivo infection rates by covalently bound furanones. Biomaterials 25(20):5023–5030. https://doi.org/10.1016/j.biomaterials.2004.01.048

    Article  CAS  PubMed  Google Scholar 

  131. Zuberi A, Misba L, Khan AU (2017) CRISPR interference (CRISPRi) inhibition of luxS gene expression in E. coli: an approach to inhibit biofilm. Front Cell Infect Microbiol 7:214. https://doi.org/10.3389/fcimb.2017.00214

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Chew SC, Yang L (2015) Biofilms. In: Caballero B, Finglas PM, Toldrá F (eds) Encyclopedia of food and health. Elsevier, Netherlands, pp 407–415. https://doi.org/10.1016/b978-0-12-384947-2.00069-6.

  133. Lahiri D, Nag M, Banerjee R, Mukherjee D, Garai S, Sarkar T, Dey A et al (2021) Amylases: biofilm inducer or biofilm inhibitor? Front Cell Infect Microbiol 11:660048. https://doi.org/10.3389/fcimb.2021.660048

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Oulahal N, Martial-Gros A, Bonneau M, Blum LJ (2007) Removal of meat biofilms from surfaces by ultrasounds combined with enzymes and/or a chelating agent. Innov Food Sci Emerg Technol 8(2):192–196. https://doi.org/10.1016/j.ifset.2006.10.001

    Article  CAS  Google Scholar 

  135. Klein MI, Duarte S, Xiao J, Mitra S, Foster TH, Koo H (2009) Structural and molecular basis of the role of starch and sucrose in Streptococcus mutans biofilm development. Appl Environ Microbiol 75(3):837–841. https://doi.org/10.1128/AEM.01299-08

    Article  CAS  PubMed  Google Scholar 

  136. Tetz GV, Artemenko NK, Tetz VV (2009) Effect of DNase and antibiotics on biofilm characteristics. Antimicrob Agents Chemother 53(3):1204–1209. https://doi.org/10.1128/aac.00471-08

    Article  CAS  PubMed  Google Scholar 

  137. Das T, Sharma PK, Busscher HJ, Van Der Mei HC, Krom BP (2010) Role of extracellular DNA in initial bacterial adhesion and surface aggregation. Appl Environ Microbiol 76(10):3405–3408. https://doi.org/10.1128/aem.03119-09

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Periasamy S, Joo HS, Duong AC, Bach THL, Tan VY, Chatterjee SS, Cheung GYC et al (2012) How Staphylococcus aureus biofilms develop their characteristic structure. Proc Natl Acad Sci U S A 109(4):1281–1286. https://doi.org/10.1073/pnas.1115006109

    Article  PubMed  PubMed Central  Google Scholar 

  139. Nijland R, Hall MJ, Burgess JG (2010) Dispersal of biofilms by secreted, matrix-degrading, bacterial DNase. PLoS ONE 5(12):15668. https://doi.org/10.1371/journal.pone.0015668

    Article  CAS  Google Scholar 

  140. Artini M, Papa R, Scoarughi GL, Galano E, Barbato G, Pucci P, Selan L (2013) Comparison of the action of different proteases on virulence properties related to the staphylococcal surface. J Appl Microbiol 114(1):266–277. https://doi.org/10.1111/jam.12038

    Article  CAS  PubMed  Google Scholar 

  141. Rahman MM, Inoue A, Tanaka H, Ojima T (2010) Isolation and characterization of two alginate lyase isozymes, AkAly28 and AkAly33, from the common sea hare Aplysia kurodai. Comp Biochem Physiol B Biochem Mol Biol 157(4):317–325. https://doi.org/10.1016/j.cbpb.2010.07.006

    Article  CAS  PubMed  Google Scholar 

  142. Singh RP, Gupta V, Kumari P, Kumar M, Reddy CRK, Prasad K, Jha B (2011) Purification and partial characterization of an extracellular alginate lyase from Aspergillus oryzae isolated from brown seaweed. J Appl Phycol 23(4):755–762. https://doi.org/10.1007/s10811-010-9576-9

    Article  CAS  Google Scholar 

  143. Wang Y, Guo EW, Yu WG, Han F (2013) Purification and characterization of a new alginate lyase from a marine bacterium Vibrio sp. Biotechnol Lett 35(5):703–708. https://doi.org/10.1007/s10529-012-1134-x

    Article  CAS  PubMed  Google Scholar 

  144. Daboor SM, Raudonis R, Cohen A, Rohde JR, Cheng Z (2019) Marine bacteria, a source for the alginolytic enzyme to disrupt Pseudomonas aeruginosa biofilms. Mar Drugs 17(5):307. https://doi.org/10.3390/md17050307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Dheilly A, Soum-Soutera E, Klein GL, Bazire A, Compère C, Haras D, Dufour A (2010) Antibiofilm activity of the marine bacterium Pseudoalteromonas sp. strain 3J6. Appl Environ Microbiol 76(11):3452–3461. https://doi.org/10.1128/aem.02632-09

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Dufourcq R, Chalkiadakis E, Fauchon M, Deslandes E, Kerjean V, Chanteau S, Petit E et al (2014) Isolation and partial characterization of bacteria (Pseudoalteromonas sp.) with potential antibacterial activity from a marine coastal environment from New Caledonia. Lett Appl Microbiol 58(2):102–108. https://doi.org/10.1111/lam.12162

    Article  CAS  PubMed  Google Scholar 

  147. Kalpana BJ, Aarthy S, Pandian SK (2012) Antibiofilm activity of α-amylase from Bacillus subtilis S8–18 against biofilm forming human bacterial pathogens. Appl Biochem Biotechnol 167(6):1778–1794. https://doi.org/10.1007/s12010-011-9526-2

    Article  CAS  PubMed  Google Scholar 

  148. Craigen B, Dashiff A, Kadouri DE (2011) The use of commercially available alpha-amylase compounds to inhibit and remove Staphylococcus aureus biofilms. Open Microbiol J 5:21–31. https://doi.org/10.2174/1874285801105010021

    Article  PubMed  PubMed Central  Google Scholar 

  149. Divakaran D, Chandran A, Chandran RP (2011) Comparative study on the production of α-amylase from Bacillus licheniformis strains. Braz J Microbiol 42(4):1397–1404. https://doi.org/10.1590/S1517-838220110004000022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Hatanaka K, Sugiura M (1993) Method for removing slime. Japanese Patent JP06306693A.

  151. Wiatr CL (1990) Application of cellulase to control industrial slime. US Patent 4,936,994, 13 Mar 1989.

  152. Taraszkiewicz A, Fila G, Grinholc M, Nakonieczna J (2013) Innovative strategies to overcome biofilm resistance. Biomed Res Int 2013:150653. https://doi.org/10.1155/2013/150653

    Article  CAS  PubMed  Google Scholar 

  153. Ghosh A, Jayaraman N, Chatterji D (2020) Small-molecule inhibition of bacterial biofilm. ACS Omega 5(7):3108–3115. https://doi.org/10.1021/acsomega.9b03695

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Gupta R, Gigras P, Mohapatra H, Goswami VK, Chauhan B (2003) Microbial α-amylases: a biotechnological perspective. Process Biochem 38(11):1599–1616. https://doi.org/10.1016/S0032-9592(03)00053-0

    Article  CAS  Google Scholar 

  155. Stutz AE, Wrodnigg TM (2011) Imino sugars and glycosyl hydrolases: historical context, current aspects, emerging trends. Adv Carbohydr Chem Biochem 66:187–298. https://doi.org/10.1016/B978-0-12-385518-3.00004-3

    Article  CAS  PubMed  Google Scholar 

  156. Saboury AA (2002) Stability, activity and binding properties study of α-amylase upon interaction with Ca2+ and Co2+. Biologia 57(11):221–228

    CAS  Google Scholar 

  157. Toda H, Nitta Y, Asanami S, Kim JP, Sakiyama F (1993) Sweet potato β-amylase: primary structure and identification of the active-site glutamyl residue. Eur J Biochem 216(1):25–38. https://doi.org/10.1111/j.1432-1033.1993.tb18112.x

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I would like to thank Marian Centre for Advanced Research, St. Mary’s College Thrissur, Kerala, India, and Authorities of St. Thomas College Palai, Kottayam, Kerala, India, for providing all necessary support.

Funding

This work was not supported by any funding agency.

Author information

Authors and Affiliations

Authors

Contributions

Dr. KV conceived the idea and outlined the content; Ms.SV collected information, reviewed the literature and developed the manuscript; Dr. KV and Dr. BM proofread and edited the manuscript. All authors read and approved the final manuscript for submission.

Corresponding author

Correspondence to Bince Mani.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Informed consent for publication of image within the text [Ghosh A, Jayaraman N, Chatterji D (2020) Small-Molecule Inhibition of Bacterial Biofilm. ACS Omega 5(7):3108–3115. Direct link: < https://pubs.acs.org/doi/10.1021/acsomega.9b03695 >] was obtained to be published in the above journal and article. Further permission related to the material excerpted should be directed to the ACS.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vani, S., Vadakkan, K. & Mani, B. A narrative review on bacterial biofilm: its formation, clinical aspects and inhibition strategies. Futur J Pharm Sci 9, 50 (2023). https://doi.org/10.1186/s43094-023-00499-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-023-00499-9

Keywords