
Biofilms are aggregates of bacteria, in most cases, which are resistant usually to broad-spectrum antibiotics in their typical concentrations or even in higher doses. A trend of increasing multi-drug resistance in biofilms, which are responsible for emerging life-threatening nosocomial infections, is becoming a serious problem. Biofilms, however, are at various sensitivity levels to environmental factors and are versatile in infectivity depending on virulence factors. This review presents the fundamental information about biofilms: formation, antibiotic resistance, impacts on public health and alternatives to conventional approaches. Novel developments in micro-biosystems that help reveal the new treatment tools by sensing and characterization of biofilms will also be discussed. Understanding the formation, structure, physiology and properties of biofilms better helps eliminate them by the usage of appropriate antibiotics or their control by novel therapy approaches, such as anti-biofilm molecules, effective gene editing, drug-delivery systems and probiotics.
Citation: Ruba Mirghani, Tania Saba, Hebba Khaliq, Jennifer Mitchell, Lan Do, Liz Chambi, Kelly Diaz, Taylor Kennedy, Katia Alkassab, Thuhue Huynh, Mohamed Elmi, Jennifer Martinez, Suad Sawan, Girdhari Rijal. Biofilms: Formation, drug resistance and alternatives to conventional approaches[J]. AIMS Microbiology, 2022, 8(3): 239-277. doi: 10.3934/microbiol.2022019
[1] | Wei Zhang, Ai Ma, Aseel Takshe, Bishr Muhamed Muwafak . Establishment of differential model of recovery treatment for children with minor brain injury and mental disorder syndrome. Mathematical Biosciences and Engineering, 2021, 18(5): 5614-5624. doi: 10.3934/mbe.2021283 |
[2] | Xiaodong Zhu, Liehui Jiang, Zeng Chen . Cross-platform binary code similarity detection based on NMT and graph embedding. Mathematical Biosciences and Engineering, 2021, 18(4): 4528-4551. doi: 10.3934/mbe.2021230 |
[3] | Mohamad Al Bannoud, Tiago Dias Martins, Silmara Aparecida de Lima Montalvão, Joyce Maria Annichino-Bizzacchi, Rubens Maciel Filho, Maria Regina Wolf Maciel . Integrating biomarkers for hemostatic disorders into computational models of blood clot formation: A systematic review. Mathematical Biosciences and Engineering, 2024, 21(12): 7707-7739. doi: 10.3934/mbe.2024339 |
[4] | Junji Ito, Emanuele Lucrezia, Günther Palm, Sonja Grün . Detection and evaluation of bursts in terms of novelty and surprise. Mathematical Biosciences and Engineering, 2019, 16(6): 6990-7008. doi: 10.3934/mbe.2019351 |
[5] | Yu Jin, Zhe Ren, Wenjie Wang, Yulei Zhang, Liang Zhou, Xufeng Yao, Tao Wu . Classification of Alzheimer's disease using robust TabNet neural networks on genetic data. Mathematical Biosciences and Engineering, 2023, 20(5): 8358-8374. doi: 10.3934/mbe.2023366 |
[6] | Jingren Niu, Qing Tan, Xiufen Zou, Suoqin Jin . Accurate prediction of glioma grades from radiomics using a multi-filter and multi-objective-based method. Mathematical Biosciences and Engineering, 2023, 20(2): 2890-2907. doi: 10.3934/mbe.2023136 |
[7] | Natalya Shakhovska, Vitaliy Yakovyna, Valentyna Chopyak . A new hybrid ensemble machine-learning model for severity risk assessment and post-COVID prediction system. Mathematical Biosciences and Engineering, 2022, 19(6): 6102-6123. doi: 10.3934/mbe.2022285 |
[8] | Marios G. Krokidis, Themis P. Exarchos, Panagiotis Vlamos . Data-driven biomarker analysis using computational omics approaches to assess neurodegenerative disease progression. Mathematical Biosciences and Engineering, 2021, 18(2): 1813-1832. doi: 10.3934/mbe.2021094 |
[9] | Lu Yu, Yuliang Lu, Yi Shen, Jun Zhao, Jiazhen Zhao . PBDiff: Neural network based program-wide diffing method for binaries. Mathematical Biosciences and Engineering, 2022, 19(3): 2774-2799. doi: 10.3934/mbe.2022127 |
[10] | Joanna Tyrcha, John Hertz . Network inference with hidden units. Mathematical Biosciences and Engineering, 2014, 11(1): 149-165. doi: 10.3934/mbe.2014.11.149 |
Biofilms are aggregates of bacteria, in most cases, which are resistant usually to broad-spectrum antibiotics in their typical concentrations or even in higher doses. A trend of increasing multi-drug resistance in biofilms, which are responsible for emerging life-threatening nosocomial infections, is becoming a serious problem. Biofilms, however, are at various sensitivity levels to environmental factors and are versatile in infectivity depending on virulence factors. This review presents the fundamental information about biofilms: formation, antibiotic resistance, impacts on public health and alternatives to conventional approaches. Novel developments in micro-biosystems that help reveal the new treatment tools by sensing and characterization of biofilms will also be discussed. Understanding the formation, structure, physiology and properties of biofilms better helps eliminate them by the usage of appropriate antibiotics or their control by novel therapy approaches, such as anti-biofilm molecules, effective gene editing, drug-delivery systems and probiotics.
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder that is diagnosed in childhood. It is characterized by impulsivity, inattention, and hyperactivity [1], which have a detrimental impact on children's learning, emotions, and social relationships. In recent years, pattern recognition has been applied to neurological disease diagnosis, and significant progress has been made in ADHD diagnosis through the develpopment of various classification approaches that utilize machine learning (ML) and deep learning (DL) techniques [2,3]. Nowadays, improving diagnosis accuracy remains a practical challenge for current ADHD classification methods, primarily due to complex factors such as limited data size and noise disturbance in sampled data. Furthermore, the identification of discriminative biomarkers for ADHD represents an essential application requirement. These biomarkers can serve as keys to uncover the mechanisms underlying ADHD and facilitate more convenient and accurate diagnosis and treatment. Accomplishing this task requires achieving satisfactory accuracy and effectively addressing the challenge of interpreting features obtained through the use of learning algorithms.
As we know, the type of biosignal is crucial in ADHD classification and biomarker detection. Suitable biosignals can extremely improve ADHD classification accuracy and yield highly credible biomarker results. Here, the magnetic resonance imaging (MRI) technique provides plenty of metrics, such as cortical thickness, gray matter probability, regional homogeneity (ReHo), amplitude of low-frequency fluctuation (ALFF), and functional connectivity (FC), to elucidate the brain status of patients. Voxel-level metrics describe the detailed brain state at a high spatial resolution. For example, complementary features can be extracted from voxel-level functional MRI (fMRI) and structural MRI (sMRI) data to characterize differences among subjects [4], while binary voxel-level ALFF feature maps are employed and input to an attention-based DL classification network [5]. However, the extraction of features from voxel-level data has significant problems, as learned features often suffer from noise interference due to the limited consideration of relationships among these voxel-level data. As a result, the performance of voxel-based ADHD classification methods is poor, greatly reducing the reliability of biomarker detection in these studies. Region-level metrics can be obtained by matching the original resolution fMRI data with a certain brain template, which preserves the functional similarity of the voxels within the region and effectively elevates the classification accuracy, while providing better interpretability for ADHD disease. Therefore, region-level metrics are preferred. For example, region-level FC has been frequently utilized for accurate ADHD classification and biomarker analysis [6,7,8]. However, FC is a numerical measure that assesses the correlation between different brain regions. Consequently, when utilized, the identified biomarkers are dispersed in practice [7], making it difficult to in focus on specific brain regions and undermining the interpretability of the obtained outcomes. Studies have shown that spontaneous low-frequency (< 0.08 Hz) fluctuations are highly synchronized between the motor cortices, while ALFFs imply spontaneous neuronal activity within the region [9]. Therefore, region-level ALFF has better functional aggregation, making it more suitable for the detection of biological markers of ADHD. We employed region-level ALFF for ADHD classification and biomarker detection in this study, where abnormal ALFF values serve as indicators of changes in ADHD-related brain regions.
The effectivity of biomarker detection also depends on the detection approaches. As far as we know, there exist two major ways to elucidate ADHD biomarkers. 1) One way is statistical analysis, which compares the differences between ADHD group and control group data. Here, the characteristics associated with credible brain region differences were found by various statistical methods including the volumes of hippocampus and amygdala[10] and the activity intensity of the cerebellum area[9]. Unfortunately, these statistical conclusions have limited capacity in ADHD individual diagnosis, since the ADHD condition is different for each subject. 2) The other way is to employ feature selection or learning strategies during the ADHD classification procedure. For example, high-score FC vectors are determined using by a specific feature ranking method[6], and the learned convolution kernels are mapped to three-dimensional space via reconstruction techniques to determine the location of highlighted brain regions[11]. But, it is highly challenging to design a satisfactory classification method that is also capable of reliable biomarker detection. On one hand, existing classification methods with ML are good at employing feature selection to find typical features from the input raw data through the various effective algorithms, such as a support vector machine with recursive feature elimination (SVM-RFE) [12], the least absolute shrinkage and selection operator (LASSO) [13,14], and the elastic net [15]. However, ML-based methods frequently use linear classifiers for ADHD prediction, and they cannot fully describe the complex relationship between features and ADHD disease. Consequently, it has low accuracy [16] and does not provide reliable biomarkers. On the other hand, DL methods exhibit remarkable classification performance, as their flexible feature learning capability facilitates the learning of potential ADHD-related features. For example, three-dimensional [4] and four-dimensional [17] convolutional neural networks (CNNs) have been directly applied to sMRI and fMRI medical image data, exploring the time and spatial patterns of ADHD features in MRI data. Several attention mechanisms have been adopted to adaptively emphasize the learned discriminative features and thus improve the classification performance [18,19]. In addition, autoencoder (AE) networks have also been widely used in ADHD classification. A deep variational AE (DVAE) network and its advanced version, the spatiotemporal attention AE (STAAE) network, have yielded impressive classification results with an accuracy of over 93% [20,21]. However, in these cases, the learned features from DL-based methods are viewed as high-level features, which have poor interpretability and cannot meet the biomarker detection requirement. In summary, both methods for biomarker detection have limitations. However, it is an interesting idea to combine the two, as it would introduce the prior knowledge obtained from statistical analysis into the classification method to guide the classification and enhance the interpretability of related derived biomarkers.
Attracted by the superior performance of the AENet-based binary hypothesis testing (BHT) framework, we have employed this model to elucidate the related biomarkers from regional ALFF data. The main contributions of the study are described as follows:
1) We use prior knowledge to guide the classification procedure. Since ADHD children suffer in executive functions, cognitive and emotional control, we applied ALFF to the data on the 50 related brain regions in the limbic system and cerebellum. Moreover, using the attributes of ALFFs as a basis, we modified the existing AENet to be more suitable for feature learning on ALFF data. Considering that the ALFF data on limited brain regions cause the AENet to exhibit unstable feature learning and thus degenerate the classification performance, we have also introduced an ensemble learning strategy to enhance the accuracy. As a result, we achieved remarkable results on multiple datasets with an average accuracy of 93.3%.
2) Regarding high-level classification performance, biomarker detection was effectively carried out. Several potential biomarkers were identified from the selected features by implementing an SVM-RFE algorithm in the BHT framework. Then, we employed a two-sample t-test between groups and correlation analysis based on the ADHD symptom scores to verify the rationality of these biomarkers. We found that brain regions, including the thalamus, hippocampus, and cerebellar lobule Ⅹ, were most discriminative in our experiments, which is in line with the existing statistical analysis reports. It further demonstrates that the biomarkers obtained via classification and statistical analyses exhibit consistency in the limbic system and cerebellum.
The limbic system involves a group of regions in the paleocortex that support various functions related to emotion regulation and motivation [22]. In detail, it mainly consists of the amygdala, hippocampus, striatum, and thalamus, which are known to be implicated in ADHD. Several studies have confirmed significant statistical differences between ADHD and healthy control (HC) subjects in the limbic system. For example, structural integrity is impaired in brain regions such as the thalamus [23], caudate nucleus [24], and amygdala [25,26] in children with ADHD. Additionally, abnormal volumes have been observed in these above regions, as well as the hippocampus [27,28]. Moreover, FC analysis has revealed significant variations in neural connections involving the thalamus and hippocampus regions [29,30].
Initially believed to be primarily involved in motor learning and coordination, the cerebellum is now recognized to play a significant role in cognition and emotion, thus making a vital contribution to the pathophysiology of ADHD [31]. Numerous studies have shown that the cerebellum undergoes the most noticeable structural and functional changes for ADHD [32]. Children with ADHD also exhibit a significant decrease in the ALFF signal in the bilateral cerebellum [9], accompanied by impaired structural integrity [33,34].
In summary, statistical differences associated with ADHD have been found in these brain regions, which means that these findings highlight the potential of the limbic system and cerebellum to serve as a source of biomarkers for ADHD detection and assist in its classification process. Hence, using this prior knowledge, the limbic system and cerebellum can be used as input brain regions. In this way, highly correlated input data can be obtained, and the interpretability of the intermediate features generated via the classification method is also enhanced.
To the best of our knowledge, two commonly employed classification frameworks exist in the field of ADHD diagnosis: the training-test framework and the hypothesis-test framework [7]. The training-test framework dominates ADHD classification, involving the learning of features from training data their comparison with those of test data to predict labels. However, this strategy proves inadequate for small-sized datasets, leading to unsatisfactory performance. These learned training-data features fail to fully encompass the characteristics of test data, resulting in a significant hindrance to accuracy improvement due to the limitation of sample number[7]. In contrast, the hypothesis-test framework follows a semi-supervised approach, where we have successfully designed a BHT framework for ADHD classification [35,36,37]. The core idea behind the BHT framework is to incorporate the test data, along with an assumed label, into the feature learning process for the training data. In scenarios in which the assumed label is incorrect, this feature learning process becomes disrupted because the test data introduces noise-like elements that are not present in the training data. As a result of comparing the training features learned under different assumptions, predicting the labels of the test data becomes easier. Therefore, the BHT framework is suitable for research on the ADHD-200 dataset due to its anti-noise ability and ability to overcome the limitation of sample size.
Figure 1 depicts the flowchart of the existing BHT framework, which utilizes FC as the raw data input (more details can be found in reference [7]). The test data were initially categorized as either HC (H0) or ADHD (H1) data. For feature selection, the SVM-RFE approach is employed to choose the N most characteristic connections for both the training and test data, wherein only the connections from the training data are retained to form two typical feature sets, XH0 and XH1. Subsequently, a feature extraction process is performed by using a modified AE network to process these feature sets and generate the corresponding high-level feature sets, ˜XH0 and ˜XH1. During the ADHD decision step, the inter-class and intra-class variability scores, DH0 and DH1, are computed for the high-level feature sets and compared. The assumption with a smaller variability score is identified as the true hypothesis, ˜Htrue, with the corresponding label assigned to the test data.
It should be noted that this FC-based model suffers from two problems in biomarker detection. Firstly, during feature selection, a total of 4005 connections are used, resulting in scattered characteristic connections. This severely undermines the meaningfulness of the identified biomarkers, as we believe that biomarkers should be localized in specific regions. Second, no prior knowledge is considered during feature selection. As a result, this purely data-driven framework may introduce bias and compromise the reliability of the biomarkers. To overcome these problems, we propose using a limited number of regional ALFFs in the limbic system and cerebellum for biomarker detection.
All the data used in this study were taken from the Athena pipeline in the ADHD-200 preprocessed dataset of the (http://preprocessed-connectomes-project.org/adhd200/), which contains data of seven sites from the ADHD-200 competition dataset. Note that the University of Pittsburgh and Washington University in sT. Louis sites only contain HC samples, while there is relatively little research on the Oregon Health & Science University sites. Thus, we ultimately chose the remaining four sites for the experiment, including New York University Medical Center (NYU), Peking University (PU, comprising three PU sub-sites), Kennedy Krieger Institute (KKI), and NeuroIMAGE (NI). Detailed information on the samples from these four datasets is recorded in Table 1.
Site | Age | Female/Male | Control | ADHD | Total |
NYU | 7–18 | 76/140 | 98 | 118 | 216 |
PU | 8–17 | 52/142 | 116 | 78 | 194 |
KKI | 8–13 | 37/46 | 61 | 22 | 83 |
NI | 11–22 | 17/31 | 23 | 25 | 48 |
We utilized a meticulous approach to extract regional ALFFs from the downloaded time course values of blood-oxygen-level-dependent (BOLD) signals recorded from the subjects in our study. To derive the voxel-based ALFFs, we implemented several standard operations, including filtering (band-pass 0.009–0.08 Hz) to eliminate noise, performing a fast Fourier transform, and calculating the power spectrum proportionally. Subsequently, we employed the automated anatomical labeling (AAL-116) atlas to divide the brain into distinct regions. Finally, regional ALFF values were obtained by averaging the voxel-based ALFF values within the regions of interest (ROIs), where a total of 50 ROIs are provided in Table 2 with their brain indices. But please note that there might be a slight deviation for the limbic system, which contains the entire cingulate gyrus including the anterior, middle, and posterior cingulate gyri.
Region name | Region index | Region name | Region index |
Olfactory cortex | 21–22 | Amygdala | 41–42 |
Insula | 29–30 | Caudate | 71–72 |
Anterior cingulate | 31–32 | Putamen | 73–74 |
Middle cingulate | 33–34 | Pallidum | 75–76 |
Posterior cingulate | 35–36 | Thalamus | 77–78 |
Hippocampus | 37–38 | Cerebellum | 91–108 |
Parahippocampal | 39–40 | Vermis | 109–116 |
We introduce the ALFF-based BHT framework for ADHD classification, wherein regional ALFFs replace FC data as the input raw data. Similar to the FC-based framework, we focus on enhancing the feature extraction step within the utilized AE network. The architecture of our ALFF-based AE network is illustrated in Figure 2. Within this diagram, the typical features (i.e., characteristic ALFF) of the training data are shown to undergo encoding via an encoder subnetwork to derive their high-level representations. These representations are then passed through a decoder subnetwork for reconstruction. Simultaneously, a classification subnetwork supervises the labeling of high-level features acquired through the encoder subnetwork. This strategy aims to retain category information related to the training data within the high-level features.
In contrast to the FC-based AE network [7], we have introduced several targeted modifications to our network architecture, making it specifically tailored to harness the attributes of ALFF. Notably, we have incorporated a subsequent ReLU into the dense layer within the decoding subnetwork. This adjustment ensures that the reconstructed ALFF maintains non-negative values. Furthermore, in practical implementation, our AE network employs the ALFF-based selected features with an output dimension (i.e., the output dimension of SVM-RFE) that has been reduced to 25, as opposed to the 50 dimensions pf the FC-based features. This conscious reduction mitigates the risk of overfitting that might arise when directly inputting these features into the FC-based AE network. Consequently, we have optimized the architecture of the classification subnetwork. Presently, the classification subnetwork employs just two residual network (ResNet) blocks, designed to process an input vector with dimensions of 8 × 1. This departure from the 20 × 1 input used in the FC-based AE network results in the generation of more efficacious high-level features. Lastly, we have meticulously detailed the parameters for each dense unit within our AE network in Table 3, note that the size of each dense layer is determined by grid search. The loss functions applied for the reconstructing of the selected features and prediction of the corresponding labels closely align with those employed in a previous study [7], ensuring methodological consistency.
Layer size* | |
Dense 1 | (25, 10) |
Dense 2 | (10, 25) |
Dense 3 | (10, 8) |
Dense 4 | (8, 8) |
Dense 5 | (8, 2) |
* The parameters (a, b) describe a dense unit with input size of a and output size of b. |
Although adjusting the AE network to accommodate ALFF data represents a positive step, a prominent challenge persists. It centers around the inherent instability of the AE network, which directly impacts the final prediction outcomes. Throughout network training, the initial configuration of learned parameters wields substantial influence. Optimal initial values possess the capacity to expedite the training process and guide the network toward a state of stability. However, when handling a small-sized dataset, the effect of these initial values is disproportionately amplified, thereby compromising the network's robustness and introducing an element of uncertainty. In practical implementation, our ALFF-based AE network is susceptible to this uncertainty. Even when holding the ALFF input constant for both training and test data, the resulting ADHD prediction outcomes may exhibit minor variances with a low probability. This uncertainty significantly impedes the pursuit of refined ADHD classification accuracy, consequently undermining the efficacy of biomarker detection endeavors.
In this study, we have employed ensemble learning as a powerful tool to address this challenge. Ensemble learning stands as a classical strategy that is renowned for mitigating issues related to data imbalance, model robustness, and uncertainty estimation. Its basic idea is to integrate several weak classifiers and build a fortified classifier, which engenders heightened reliability within the classification outcomes. The existing examples involve the collection of classifiers from the fields of ML and DL for application in various medical diagnostic scenarios, such as Parkinson's disease classification [38], anticancer peptide prediction [39], and virulence factor detection [40]. This strategy was also successfully utilized for ADHD diagnosis, where solid decision-making can be realized based on multimodal data [41].
The proposed ensemble classification method is illustrated in Figure 3. For a given test data, the process involves generating the typical features sets (XH0 and XH1) of training data based on opposite test label assumptions in the feature selection step. Then, multiple pairs of AE networks are utilized, each initialized with random parameters, to acquire multiple pairs of high-level feature sets, ˜XH0 and ˜XH1. Subsequently, pairwise high-level features sets are compared to determine the true hypothesis and corresponding hypothesis labels (Ltrue), which compares the intra-class and inter-class distances ratios of each feature set. The hypothesis label is considered as the prediction result of a baseline classifier. Finally, a hard voting strategy is then applied to these hypothesis labels. By assessing the frequency of label values (0 for ADHD and 1 for HC), class labels of the data are assigned higher frequency label values.
As described for the BHT framework in Section 2.2, typical feature sets were selected by the SVM-RFE algorithm in the feature selection stage. Meanwhile, reliability weights Wji of typical feature sets can be obtained during this process. Specifically, the SVM-RFE algorithm trains a linear SVM in one iteration to obtain a weight vector that fits to the input feature set. Then, the square of the weight vector is used as the criterion for judging the usefulness of features, and the feature with the smallest-valued criterion is removed. After removing a certain number of features over multiple iterations, a typical feature set with the expected dimensions is obtained. The square of the fitted linear SVM weight vector on this feature set is considered to comprise the reliability weights of the features.
Reliability weights not only measure the contribution of features to classification they also serve as a criterion for subsequently extracting brain biomarkers. However, there are differences in the values and ranks of reliability weights at different sites. Moreover, under the binary hypothesis framework, the typical feature subset generated under the correct hypothesis is more valuable. Therefore, the weighted average of the feature reliability weights were designed to be from the correct hypotheses for the four sites, where the weighted value is the product of the number of people and the classification accuracy for the site. The weighted average result is seen as a more comprehensive measure to evaluate each feature, and it is called the feature score Si for each ROI. The feature score can be defined as follows:
Si=∑4j=0Accj×Nj×Wji∑4j=0Accj×Nj, | (3.1) |
where Accj and Nj respectively denote the classification accuracy after ensemble learning and data size of the j-th dataset, Wji represents the reliability weight for the i-th ROI of the j-th dataset. This feature score provides enough convenience for finding ADHD biomarkers.
The classification performance of our BHT framework with ALFF-based AE network is presented in Table 4, where the accuracies with and without ensemble learning are provided. Specifically, each site was subjected to 50 leave-one-out cross-validation (LOOCV) trials, and the average accuracy without ensemble learning strategies was obtained. The results with ensemble learning were derived from the average of 1000 hard voting trials, where each trial entailed randomly selecting seven hypothesis labels (Ltrue) from the 50 LOOCV trials. During the AE network learning, an Adam optimizer is utilized to optimize the whole network. Hyperparameters of each site were determined through grid search, including the learning rate, training epoch, and the rate of dropout. After a certain number of epochs, the training loss converges and becomes stable. In addition, ablation experiments were designed to verify the role of components of the ALFF-based AE network, as well as under the BHT framework. 1) Default-classifier network: the classifier subnetwork was hidden, only the complete AE network was retained, and the entire network was trained by using the reconstruction loss. 2) Default-decoder network: the decoder subnetwork was hidden, the encoder degenerated into a dense layer, and the whole network was trained by using the cross-entropy loss. Later, ADHD decisions were made based on the encoder output features of the default-classifier network and the output features of the first dense layer of the default-decoder network, respectively. The results of ablation experiments are shown in Table 5, which shows that the hyperparameter and experimental details were identical to those of the ALFF-based AE network. The source code is available at https://github.com/BiolabHHU/ALFF-based-BHT.
Site | Accuracy (%) | Sensitivity (%) | Specificity (%) | AUROC | AUPRC | MCC | F1 |
Without ensemble learning | |||||||
NYU | 91.94 | 90.59 | 93.55 | 0.9207 | 0.8913 | 0.8396 | 0.9239 |
PU | 80.32 | 78.87 | 81.29 | 0.8008 | 0.7984 | 0.5994 | 0.7697 |
KKI | 88.19 | 87.45 | 88.46 | 0.8796 | 0.8817 | 0.7445 | 0.8256 |
NI | 87.38 | 88.40 | 86.26 | 0.8733 | 0.8296 | 0.7488 | 0.8786 |
Average | 86.96 | 86.33 | 87.39 | 0.8686 | 0.8503 | 0.7331 | 0.8495 |
With ensemble learning | |||||||
NYU | 96.20 | 96.88 | 95.38 | 0.9616 | 0.9400 | 0.9250 | 0.9660 |
PU | 88.79 | 96.67 | 83.50 | 0.8982 | 0.9074 | 0.7801 | 0.8719 |
KKI | 95.05 | 98.90 | 93.66 | 0.9656 | 0.9815 | 0.9033 | 0.9276 |
NI | 93.23 | 98.32 | 87.70 | 0.9302 | 0.9174 | 0.8667 | 0.9374 |
Average | 93.32 | 97.69 | 90.06 | 0.9389 | 0.9366 | 0.8688 | 0.9257 |
Network | NYU (%) | PU (%) | KKI (%) | NI (%) | Average (%) |
Without ensemble learning | |||||
Default-classifier network | 69.00 | 53.69 | 42.82 | 67.67 | 58.29 |
Default-classifier network | 89.83 | 80.26 | 86.84 | 86.79 | 85.93 |
ALFF-based AE network | 91.94 | 80.32 | 88.19 | 87.38 | 86.86 |
With ensemble learning | |||||
Default-classifier network | 75.01 | 52.38 | 35.51 | 75.49 | 59.60 |
Default-classifier network | 95.14 | 85.45 | 91.29 | 92.39 | 91.07 |
ALFF-based AE network | 96.20 | 88.79 | 95.05 | 93.23 | 93.32 |
In Table 4, one can see that an evident distinction emerges in the classification performance when comparing the results in the absence of ensemble learning to those in its presence, and it is manifested in the differences in metrics, including accuracy, sensitivity, specificity, area under the receiver operating characteristic curve (AUROC), area under the precision-recall curve (AUPRC), F1 score (F1), and apply capitalization correlation coefficient (MCC). The absence of ensemble learning resulted in notably inferior performance, as indicated by the average accuracy of 87.0%. Notably, when confronted with the PU dataset, the accuracy decreased to an unsatisfactory 80.3%. This decline is attributed to a high prevalence of comorbidity disorders among the ADHD children within this dataset, affecting 44 out of 78 cases. Conversely, the application of ensemble learning resulted in a marked enhancement in the accuracy metrics. The average accuracy significantly increased, reaching 93.3%. This positive impact is particularly evident in the case of the PU dataset, where accuracy improved dramatically from the aforementioned 80.3% to 88.8%. This result effectively substantiates the ability of ensemble learning to augment the potency of weaker classifiers and make them robust. Moreover, Table 4 also presents the results of measurements of the area under the curve. Specifically, the average AUROC and AUPRC values were both around 0.93, underscoring the excellent performance of the ALFF-based BHT framework on the ADHD-200 dataset. Remarkably, this heightened level of accuracy was achieved through the utilization of a mere 25 selected features from 50 regional ALFFs. This outcome serves to reinforce the presumption that our approach facilitates the biomarker detection endeavors. Additionally, we provide the receiver operating characteristic (ROC) curves for the used datasets in Figure 4, which shows that the curve for the KKI site had the largest area among these datasets.
To further validate the utilization of ensemble learning, we constructed a box-and-whisker plot to show the accuracy distribution, as shown in Figure 5. It is evident that the adoption of ensemble learning leads to an enhancement in the average accuracy. While this learning strategy may not entirely eliminate the fluctuations in accuracy owing to the inherent uncertainty of the AE network, can be ascertained from Table 4, there was a substantial reduction in the standard deviation of accuracy. Especially, the enhancement of accuracy are obviously disclosed on the PU dataset. These findings prove the effectiveness of ensemble learning as a tool to enhance the robustness of our ALFF-based BHT framework.
Table 5 shows a comparison of the classification accuracy of the three networks. The accuracy of the default-classifier network was only 42–76%, which may have been caused by the lack of information learned from the label based on the guidance of the cross-entropy loss; thus, the extracted high-level features had no inter-class discrimination. The classification accuracy of the default-decoder network was 1–4% lower that of proposed ALFF-based AE network, indicating that the lack of reconstruction loss may reduce the representation ability of high-level features.
We tested our method against other state-of-the-art methods the results are presented in Table 6. The compared methods include ML-based methods such as R-Relief [42], L1BioSVM [43], and Fusion fMRI [44], as well as DL-based methods such as the 3D CNN [4], DeepfMRI [45], CDAE [46], DVAE [20], STAAE [21] and data augmentation [47]. In addition, our previous work under the BH framework is also included in this comparison. These methods are referred to as SP-BH [37] and SP-l2,1-BH [36], which use subspace learning and l2,1-norm subspace learning for ADHD classification, respectively.
NYU(%) | PU(%) | KKI(%) | NI(%) | Average(%) | Material | Rawfeatures | Selectedfeatures* | Biomarkerdetection | |
ML | |||||||||
Fusion fMRI (2018) | 52.7 | - | 86.7 | 72.9 | 70.8 | FC | 4005 | - | No |
L1BioSVM (2018) | - | 81.1 | 81.3 | - | 81.2 | FC | 6670 | - | No |
R-Relief (2019) | 70.7 | 68.6 | 81.8 | 76.0 | 74.3 | fALFF | 31 × 37 × 31 | - | No |
DL | |||||||||
3D CNN (2017) | 70.5 | 63.0 | - | 72.8 | 68.8 | MRI image | 90 × 117 × 100 | 1024 | No |
DeepfMRI (2020) | 73.1 | 62.7 | - | 67.9 | 67.9 | BOLD | 90 | 32 | No |
CDAE (2021) | 73.2 | 70.6 | 81.7 | 79.0 | 76.1 | MRI image | 60 × 72× 60 | 5 × 6 × 5 | No |
DVAE (2021) | 62.4 | 67.0 | 78.1 | 68.8 | 69.1 | BOLD | 28,546 | 80 | No |
STAAE (2022) | 93.5 | 92.7 | 90.4 | 91.7 | 92.1 | BOLD | 28,546 | 100 | Yes |
Data augmentation (2023) | 75.6 | 76.5 | 76.0 | - | 76.0 | FC | 13,456 | 1856 | No |
BHT framework | |||||||||
SP-BH (2019) | 96.2 | 95.8 | 86.7 | 91.6 | 92.6 | FC | 4005 | 100 | No |
SP-l2,1-BH (2020) | 99.5 | 96.3 | 100 | 95.8 | 97.9 | FC | 4005 | 50 | No |
AENet (2022) | 99.8 | 99.6 | 99.8 | 99.3 | 99.6 | FC | 4005 | 50 | Yes |
Our | 96.2 | 88.8 | 95.1 | 93.3 | 93.3 | ALFF | 50 | 25 | Yes |
* The number of feature selected in the ML method is not fixed. And the DL method selects the learned high-level features, while the BHT method selects typical features from the raw features. |
From Table 6, it is observed that the existing ML methods had the lowest accuracies, stemming from their limited exploration of potential features for classification. Meanwhile, most examined DL methods are confined to the traditional training-test framework, which causes the learned features from the training data to inadequately representing the features of the test data. Among these methods, the STAAE approach stands out because of its remarkable performance achievements and a commendable biomarker detection capability. Its success benefits from the integration of an attention module that captures temporal patterns from fMRI spatiotemporal features. Based on its accurate classification, STAAE extends its application by projecting these temporal patterns onto corresponding brain regions, effectively fulfilling the biomarker detection task. The listed methods within the BHT framework all exhibited impressive classification performance, facilitated by the utilization of test data information (without seeing its label). Among these methods, AENet leverages DL to effectively represent high-level features, making it superior to the SP-BH, SP-l2,1-BH approaches. As an FC-based BHT method, AENet incorporates a great number of connections, consequently leading to a widespread selection of features across the entire brain. This, however, renders the identified biomarkers indiscriminate. In contrast, our ALFF-based method achieved an average accuracy of 93.3%, even though it was slightly inferior to that of AENet. A distinctive trait lies in the substantial reduction of both the used raw and selected features. This implies that the features used and selected by our algorithm carry more significance as potential ADHD biomarkers. Moreover, the selected features were tightly localized within the limbic system and cerebellum, rendering our ALFF-based method exceedingly interpretable.
Table 7 presents the top 10 brain regions obtained with the highest feature scores Si; their locations are visualized in Figure 6. These identified regions are considered to be potential ADHD biomarkers. To substantiate our findings, we conducted a correlation analysis between the ALFF data from these regions and the symptom scores provided by the ADHD-200 dataset; the results are shown in Table 7. Since the NI site did not give symptom assessment data, we performed correlation analysis for the PU, KKI and NYU sites. Notably, only individuals with ADHD-simplex were included in these analyses to mitigate the influence of comorbid disorders. In Table 7, it is evident that the majority of detected biomarkers had a robust correlation with symptom scores from the PU dataset, demonstrating significant values above 0.2, under the 95% confidence interval. This finding underscores the potential of our identified biomarkers to capture ADHD-related characteristics. However, an intriguing observation arose from the results obtained for the other two datasets. In this case, the correlation between biomarkers and symptoms was notably weaker, especially for the NYU site, as reflected by the higher P values over 0.05. This discrepancy can be attributed to the utilization of distinct symptom assessment measures across these datasets. To elaborate, although three datasets were derive from symptom measures based on questionnaires administered to parents and teachers, the specific scales employed differ. The PU dataset employs the ADHD Rating Scale-Ⅳ (ADHD-RS), whereas the KKI and NYU datasets utilize Conners' Parent Rating Scale-Revised, Long version (CPRS-LV). Despite the shared origin in parental and teacher reports, we posit that ADHD-RS offers advantages in terms of its ability to abnormalities within the limbic system and cerebellum. This assertion underscores the potential for ADHD-RS to provide insights into these specific regions.
Brian region | Ranking order | PU (ADHD-RS) | KKI (CPRS-LV) | NYU (CPRS-LV) | ||||
Name | Abbrev | Corr | P-value | Corr | P-value | Corr | P-value | |
Middle cingulate gyrus (R) | DCG.R | 1 | 0.235 | 0.007 | 0.358 | 0.003 | -0.035 | 0.660 |
Cerebellum Ⅸ (R) | CRBL9.R | 2 | 0.161 | 0.065 | 0.155 | 0.213 | 0.038 | 0.630 |
Amygdala (R) | AMYG.R | 3 | 0.198 | 0.023 | 0.057 | 0.648 | -0.005 | 0.953 |
Thalamus (L) | THA.L | 4 | 0.245 | 0.005 | 0.150 | 0.229 | 0.044 | 0.572 |
Cerebellum Ⅹ (R) | CRBL10.R | 5 | 0.244 | 0.005 | 0.241 | 0.051 | 0.021 | 0.787 |
Cerebellum Crus Ⅱ (L) | CRBL Crus2.L | 6 | 0.245 | 0.005 | 0.259 | 0.036 | 0.128 | 0.102 |
Cerebellum Crus Ⅱ (R) | CRBL Crus2.R | 7 | 0.214 | 0.014 | 0.236 | 0.057 | 0.123 | 0.115 |
Thalamus (R) | THA.R | 8 | 0.270 | 0.002 | 0.184 | 0.139 | 0.043 | 0.580 |
Hippocampus (L) | HIP.L | 9 | 0.274 | 0.001 | 0.273 | 0.027 | 0.021 | 0.786 |
Caudate (R) | CAU.L | 10 | 0.235 | 0.007 | 0.215 | 0.083 | 0.034 | 0.667 |
Figure 7 supplements our analysis and visualizes the results of correlation analysis for ADHD-RS scales, encompassing regions the left hippocampal gyrus, left and right thalamus, and left cerebellum Crus Ⅱ gyri, which are the four regions with the highest correlation coefficients. It further supports that these regions exhibit significant relevance to ADHD, thereby warranting consideration as potential biomarkers.
We shall present some biological explanations for our biomarkers. Table 7 presents the findings, highlighting significant associations within regions such as the amygdala, caudate nucleus, hippocampus, and thalamus gyri. These regions are well-established in their role in shaping and comprehending human emotions, as extensively documented in the existing literature [48,49]. Notably, ADHD patients have consistently exhibited reduced amygdala and hippocampal volumes in anatomical control experiments. Moreover, developmental delays and degenerative changes within these regions have been unveiled through lifespan exploratory modeling [10]. Concurrently, the caudate nucleus has emerged as a consistent focal point, with volumetric differences consistently noted in ADHD research [50,51,52]. Particularly, a volume asymmetry analysis demonstrated noteworthy links between caudate asymmetry and cumulative severity ratings of inattentive behaviors in ADHD-afflicted children [24]. As we delve into the thalamus, its pivotal role in information transmission, regulation, and participation in cognitive and behavioral processes such as attention, emotion, and motor control cannot be understated. Morphological aberrations in the thalamus [23] have been discovered and strongly correlated with ADHD symptom scores [53], further substantiating our identified biomarkers within the limbic system.
Furthermore, our biomarkers extend to the cerebellum, particularly in the right inferior posterior lobe of the cerebellar hemisphere, encompassing cerebellum 9, 10, and Crus Ⅱ. A longitudinal case-control study revealed that ADHD participants with worse clinical outcomes exhibited a gradual decrease in overall cerebellar volume, primarily caused by abnormalities in the posterior inferior cerebellar hemisphere [54]. Moreover, a smaller volume in the cerebellar lobule Ⅹ has been found in children [55,56]. Our findings align harmoniously with these reports. The lateral hemisphere of the cerebellum has emerged as a pivotal player in executive functions [57,58], spatial cognition, and language processing [59]. Consequently, the identified abnormalities within the posterior inferior cerebellar hemisphere may indeed contribute to the executive function deficits that constitute a prominent symptomatology of ADHD.
Overall, the aforementioned evidence firmly supports the presence of our delineated ADHD biomarkers within the limbic system and cerebellum. Here, the right middle cingulate gyrus is also established as a biomarker, but there is also a consensus that it is part of the salience network. Furthermore, when it comes to the cognitive function and emotional processing of ADHD patients, the middle cingulate gyrus is considered to play an important role [60,61]. Some existing reports have confirmed that the right middle cingulate gyrus is abnormal in ADHD patients. For example, a sex difference study on the factors related to the symptoms of common mental disorders in adolescence found that more prominent symptoms of hyperactivity/inattention were associated with lower grey matter volume of the bilateral anterior and midcingulum among boys [62]. Another study showed that compared with normal children, the increased degree centrality values for the right middle cingulate gyrus indicated differences in functional network connectivity for ADHD children [63].
Our research has two limitations. First of all, biomarker detection only involves the linear relationship between features and does not take into account the nonlinear components. Our attempt to validate the discrimination of these biomarkers through a two-sample t-test experiment did not yield statistically significant results. This indicates no inter-group differences in the ALFF features of a single brain region. However, the aggregate of brain region features still contributes to the construction of excellent high-level feature space, which is due to the powerful feature extraction ability of DL. Unfortunately, the contribution here is difficult to quantify, as it involves the amount of classification information held by the brain region features and the details of non-linear fitting performed by the neural network. This issue limits further exploration of biomarkers; thus, in the future, we will attempt to explore this limitation by using other brain region features such as the voxel-based morphometry and ReHo, as well as introduce the interpretability theory of neural networks. In addition, our algorithm achieved good classification performance on limited brain regions of the limbic system and cerebellum, indicating the importance of these brain regions for ADHD. Moreover, we conducted biomarker detection, which revealed abnormal brain regions that are consistent with previously reported anatomical abnormalities. However, it may be insufficient to explore brain regions related to the biological mechanisms of ADHD solely through ALFF data. In future studies, we will consider multi-modal features for to characterize brain regions.
This study primarily combined existing statistical prior knowledge and pattern recognition methods to elucidate critical ADHD biomarkers. Specifically, the ALFF in the limbic system and cerebellum, which is highly related to ADHD in statistical analysis, was used as input for classification, contributing to reduce the range of biomarker detection. The BHT framework and ensemble learning methods were employed to ensure high accuracy in the classification results. Consequently, we performed a highly credible biomarker detection task that achieved an average accuracy of 93% on the ADHD-200 datasets. Several brain regions such as the thalamus, hippocampus, amygdala, and cerebellum Ⅸ were extracted from the results of the SVM-RFE algorithm as biomarkers. We validated them by analyzing the correlation among symptom scores. Moreover, these findings extend previous findings and align with the existing reports on the neurobiological contributions to ADHD, which demonstrates the effectiveness of our method.
The authors declare they have not used Artificial Intelligence (AI) tools in the creation of this article.
Ying Chen: Conceptualization, Methodology, Writing–original draft; Lele Wang: Conceptualization, Software, Writing–original draft; Zhixin Li: Validation, Writing–review & editing; Yibin Tang: Software, Methodology, Writing–original draft; Zhan Huan: Supervision.
This work was supported in part by the National Natural Science Foundation of China under Grants: 62201093.
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
[1] |
Silva VO, Soares LO, Silva Júnior A, et al. (2014) Biofilm formation on biotic and abiotic surfaces in the presence of antimicrobials by Escherichia coli isolates from cases of bovine mastitis. Appl Environ Microbiol 80: 6136-6145. https://doi.org/10.1128/aem.01953-14 ![]() |
[2] |
Donlan RM (2002) Biofilms: microbial life on surfaces. Emerging Infect Dis 8: 881-890. https://doi.org/10.3201/eid0809.020063 ![]() |
[3] |
Yin W, Wang Y, Liu L, et al. (2019) Biofilms: The microbial “protective clothing” in extreme environments. Int J Mol Sci 20: 3423. https://doi.org/10.3390/ijms20143423 ![]() |
[4] |
Koerdt A, Gödeke J, Berger J, et al. (2010) Crenarchaeal biofilm formation under extreme conditions. PLoS One 5: e14104. https://doi.org/10.1371/journal.pone.0014104 ![]() |
[5] |
Chen Y, Yan F, Chai Y, et al. (2013) Biocontrol of tomato wilt disease by Bacillus subtilis isolates from natural environments depends on conserved genes mediating biofilm formation. Environ Microbiol 15: 848-864. https://doi.org/10.1111/j.1462-2920.2012.02860.x ![]() |
[6] |
Van Houdt R, Michiels CW (2010) Biofilm formation and the food industry, a focus on the bacterial outer surface. J Appl Microbiol 109: 1117-1131. https://doi.org/10.1111/j.1365-2672.2010.04756.x ![]() |
[7] |
Di Martino P (2018) Extracellular polymeric substances, a key element in understanding biofilm phenotype. AIMS Microbiol 4: 274-288. https://doi.org/10.3934/microbiol.2018.2.274 ![]() |
[8] |
Decho AW, Gutierrez T (2017) Microbial extracellular polymeric substances (EPSs) in ocean systems. Front Microbiol 8. https://doi.org/10.3389/fmicb.2017.00922 ![]() |
[9] |
Flemming HC (2016) EPS-Then and Now. Microorganisms 4: 41. https://doi.org/10.3390/microorganisms4040041 ![]() |
[10] |
Kragh KN, Hutchison JB, Melaugh G, et al. (2016) Role of multicellular aggregates in biofilm formation. mBio 7: e00237-00216. https://doi.org/10.1128/mBio.00237-16 ![]() |
[11] |
Taylor M, Ross K, Bentham R (2009) Legionella, Protozoa and Biofilms: Interactions within complex microbial systems. Microbiol Ecol 58: 538-547. https://doi.org/10.1007/s00248-009-9514-z ![]() |
[12] |
de Alexandre Sebastião F, Pilarski F, Lemos MVF (2013) Composition of extracellular polymeric substances (EPS) produced by flavobacterium columnare isolated from tropical fish in Brazil. Braz J Microbiol 44: 861-864. https://doi.org/10.1590/S1517-83822013005000058 ![]() |
[13] |
McSwain BS, Irvine RL, Hausner M, et al. (2005) Composition and distribution of extracellular polymeric substances in aerobic flocs and granular sludge. Appl Environ Microbiol 71: 1051. https://doi.org/10.1128/AEM.71.2.1051-1057.2005 ![]() |
[14] |
López D, Vlamakis H, Kolter R (2010) Biofilms. Cold Spring Harbor Perspect Biol 2: a000398. https://doi.org/10.1101/cshperspect.a000398 ![]() |
[15] |
Nadell CD, Drescher K, Wingreen NS, et al. (2015) Extracellular matrix structure governs invasion resistance in bacterial biofilms. ISME J 9: 1700-1709. https://doi.org/10.1038/ismej.2014.246 ![]() |
[16] |
Davenport EK, Call DR, Beyenal H (2014) Differential protection from tobramycin by extracellular polymeric substances from Acinetobacter baumannii and Staphylococcus aureus biofilms. Antimicrob Agents Chemother 58: 4755-4761. https://doi.org/10.1128/AAC.03071-14 ![]() |
[17] |
Donlan RM (2001) Biofilm formation: A clinically relevant microbiological process. Clin Infect Dis 33: 1387-1392. https://doi.org/10.1086/322972 ![]() |
[18] |
Lewis K (2010) Persister cells. Annu Rev Microbiol 64: 357-372. https://doi.org/10.1146/annurev.micro.112408.134306 ![]() |
[19] |
Rumbaugh KP, Sauer K (2020) Biofilm dispersion. Nat Rev Microbiol 18: 571-586. https://doi.org/10.1038/s41579-020-0385-0 ![]() |
[20] |
Rice SA, Tan CH, Mikkelsen PJ, et al. (2009) The biofilm life cycle and virulence of Pseudomonas aeruginosa are dependent on a filamentous prophage. ISME J 3: 271-282. https://doi.org/10.1038/ismej.2008.109 ![]() |
[21] |
Garrett TR, Bhakoo M, Zhang Z (2008) Bacterial adhesion and biofilms on surfaces. Prog Nat Sci 18: 1049-1056. https://doi.org/10.1016/j.pnsc.2008.04.001 ![]() |
[22] |
Donlan RM (2001) Biofilms and device-associated infections. Emerg Infect Dis 7: 277-281. https://doi.org/10.3201/eid0702.010226 ![]() |
[23] |
Donlan RM, Costerton JW (2002) Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev 15: 167-193. https://doi.org/10.1128/cmr.15.2.167-193.2002 ![]() |
[24] |
Armbruster CR, Parsek MR (2018) New insight into the early stages of biofilm formation. Proc Natl Acad Sci 115: 4317. https://doi.org/10.1073/pnas.1804084115 ![]() |
[25] |
Da Cunda P, Iribarnegaray V, Papa-Ezdra R, et al. (2019) Characterization of the different stages of biofilm formation and antibiotic susceptibility in a clinical acinetobacter baumannii Strain. Microb Drug Resist 26: 569-575. https://doi.org/10.1089/mdr.2019.0145 ![]() |
[26] |
Rasamiravaka T, Labtani Q, Duez P, et al. (2015) The formation of biofilms by Pseudomonas aeruginosa: a review of the natural and synthetic compounds interfering with control mechanisms. Biomed Res Int 2015: 759348. https://doi.org/10.1155/2015/759348 ![]() |
[27] |
Koczan JM, Lenneman BR, McGrath MJ, et al. (2011) Cell surface attachment structures contribute to biofilm formation and xylem colonization by Erwinia amylovora. Appl Environ Microbiol 77: 7031-7039. https://doi.org/10.1128/AEM.05138-11 ![]() |
[28] |
Mandlik A, Swierczynski A, Das A, et al. (2008) Pili in Gram-positive bacteria: assembly, involvement in colonization and biofilm development. Trends Microbiol 16: 33-40. https://doi.org/10.1016/j.tim.2007.10.010 ![]() |
[29] |
Berne C, Ducret A, Hardy GG, et al. (2015) Adhesins involved in attachment to abiotic surfaces by Gram-Negative bacteria. Microbiol Spectrum 3: 10. https://doi.org/10.1128/microbiolspec.MB-0018-2015 ![]() |
[30] |
Dunne WM (2002) Bacterial adhesion: seen any good biofilms lately?. Clin Microbiol Rev 15: 155-166. https://doi.org/10.1128/cmr.15.2.155-166.2002 ![]() |
[31] |
Renner LD, Weibel DB (2011) Physicochemical regulation of biofilm formation. MRS Bull 36: 347-355. https://doi.org/10.1557/mrs.2011.65 ![]() |
[32] |
Hinsa SM, Espinosa-Urgel M, Ramos JL, et al. (2003) Transition from reversible to irreversible attachment during biofilm formation by Pseudomonas fluorescens WCS365 requires an ABC transporter and a large secreted protein. Mol Microbiol 49: 905-918. https://doi.org/10.1046/j.1365-2958.2003.03615.x ![]() |
[33] |
Petrova OE, Sauer K (2012) Sticky situations: key components that control bacterial surface attachment. J Bacteriol Res 194: 2413-2425. https://doi.org/10.1128/JB.00003-12 ![]() |
[34] |
Miller JK, Badawy HT, Clemons C, et al. (2012) Development of the Pseudomonas aeruginosa mushroom morphology and cavity formation by iron-starvation: a mathematical modeling study. J Theor Biol 308: 68-78. https://doi.org/10.1016/j.jtbi.2012.05.029 ![]() |
[35] |
Abebe GM (2020) The role of bacterial biofilm in antibiotic resistance and food contamination. Int J Microbiol 2020: 1705814. https://doi.org/10.1155/2020/1705814 ![]() |
[36] |
Jefferson KK (2004) What drives bacteria to produce a biofilm?. FEMS Microbiol Lett 236: 163-173. https://doi.org/10.1111/j.1574-6968.2004.tb09643.x ![]() |
[37] |
Kurmoo Y, Hook AL, Harvey D, et al. (2020) Real time monitoring of biofilm formation on coated medical devices for the reduction and interception of bacterial infections. Biomater Sci 8: 1464-1477. https://doi.org/10.1039/c9bm00875f ![]() |
[38] |
Salgar-Chaparro SJ, Lepkova K, Pojtanabuntoeng T, et al. (2020) Nutrient level determines biofilm characteristics and subsequent impact on microbial corrosion and biocide effectiveness. Appl Environ Microbiol 86: e02885-02819. https://doi.org/10.1128/AEM.02885-19 ![]() |
[39] | Wilson C, Lukowicz R, Merchant S, et al. (2017) Quantitative and qualitative assessment methods for biofilm growth: A mini-review. Res Rev J Eng Technol 6. Available from: http://www.rroij.com/open-access/quantitative-and-qualitative-assessment-methods-for-biofilm-growth-a-minireview-.pdf |
[40] |
Haney EF, Trimble MJ, Cheng JT, et al. (2018) Critical assessment of methods to quantify biofilm growth and evaluate antibiofilm activity of host defence peptides. Biomolecules 8: 29. https://doi.org/10.3390/biom8020029 ![]() |
[41] |
Kaplan JB (2010) Biofilm dispersal: mechanisms, clinical implications, and potential therapeutic uses. J Dent Res 89: 205-218. https://doi.org/10.1177/0022034509359403 ![]() |
[42] |
Rabin N, Zheng Y, Opoku-Temeng C, et al. (2015) Biofilm formation mechanisms and targets for developing antibiofilm agents. Future Med Chem 7: 493-512. https://doi.org/10.4155/fmc.15.6 ![]() |
[43] |
Sharpton TJ, Combrink L, Arnold HK, et al. (2021) Erratum to “Harnessing the gut microbiome in the fight against anthelminthic drug resistance”. Curr Opin Microbiol 53: 26-34. https://doi.org/10.1016/j.mib.2021.01.003 ![]() |
[44] |
Seth AK, Geringer MR, Hong SJ, et al. (2012) Comparative analysis of single-species and polybacterial wound biofilms using a quantitative, in vivo, rabbit ear model. PLoS One 7: e42897. https://doi.org/10.1371/journal.pone.0042897 ![]() |
[45] |
Yang L, Liu Y, Wu H, et al. (2011) Current understanding of multi-species biofilms. Int J Oral Sci 3: 74-81. https://doi.org/10.4248/IJOS11027 ![]() |
[46] |
Lohse MB, Gulati M, Johnson AD, et al. (2018) Development and regulation of single- and multi-species Candida albicans biofilms. Nat Rev Microbiol 16: 19-31. https://doi.org/10.1038/nrmicro.2017.107 ![]() |
[47] |
Lynch AS, Robertson GT (2008) Bacterial and fungal biofilm infections. Annu Rev Med 59: 415-428. https://doi.org/10.1146/annurev.med.59.110106.132000 ![]() |
[48] |
Seneviratne G, Zavahir JS, Bandara WMMS, et al. (2007) Fungal-bacterial biofilms: their development for novel biotechnological applications. World J Microbiol Biotechnol 24: 739. https://doi.org/10.1007/s11274-007-9539-8 ![]() |
[49] | Guo YS, Furrer JM, Kadilak AL, et al. (2018) Bacterial extracellular polymeric substances amplify water content variability at the pore scale. Front Environ Sci 6. https://doi.org/10.3389/fenvs.2018.00093 |
[50] | Wingender J, Neu TR, Flemming HC, et al. (1999) What are bacterial extracellular polymeric substances?. Microbial extracellular polymeric substances: Characterization, structure and function. Berlin: Springer Berlin Heidelberg 1-19. https://doi.org/10.1007/978-3-642-60147-7_1 |
[51] |
Wilking JN, Zaburdaev V, De Volder M, et al. (2013) Liquid transport facilitated by channels in Bacillus subtilis biofilms. Proc Natl Acad Sci U S A 110: 848-852. https://doi.org/10.1073/pnas.1216376110 ![]() |
[52] |
Costa OYA, Raaijmakers JM, Kuramae EE (2018) Microbial extracellular polymeric substances: ecological function and impact on soil aggregation. Front Microbiol 9: 1636-1636. https://doi.org/10.3389/fmicb.2018.01636 ![]() |
[53] |
Navarre WW, Schneewind O (1999) Surface proteins of gram-positive bacteria and mechanisms of their targeting to the cell wall envelope. Microbiol Mol Biol Rev 63: 174. https://doi.org/10.1128/MMBR.63.1.174-229.1999 ![]() |
[54] |
Davey ME, O'Toole GA (2000) Microbial biofilms: from ecology to molecular genetics. Microbiol Mol Biol Rev: MMBR 64: 847-867. https://doi.org/10.1128/mmbr.64.4.847-867.2000 ![]() |
[55] |
Hibiya K, Tsuneda S, Hirata A (2000) Formation and characteristics of nitrifying biofilm on a membrane modified with positively-charged polymer chains. Colloids Surf, B 18: 105-112. https://doi.org/10.1016/S0927-7765(99)00141-1 ![]() |
[56] |
Navada S, Knutsen MF, Bakke I, et al. (2020) Nitrifying biofilms deprived of organic carbon show higher functional resilience to increases in carbon supply. Sci Rep 10: 7121. https://doi.org/10.1038/s41598-020-64027-y ![]() |
[57] |
Hunt SM, Werner EM, Huang B, et al. (2004) Hypothesis for the role of nutrient starvation in biofilm detachment. Appl Environ Microbiol 70: 7418-7425. https://doi.org/10.1128/AEM.70.12.7418-7425.2004 ![]() |
[58] |
Maddela NR, Zhou Z, Yu Z, et al. (2018) Functional determinants of extracellular polymeric substances in membrane biofouling: Experimental evidence from pure-cultured sludge bacteria. Appl Environ Microbiol 84: e00756-00718. https://doi.org/10.1128/AEM.00756-18 ![]() |
[59] |
Stoodley P, Sauer K, Davies DG, et al. (2002) Biofilms as complex differentiated communities. Annu Rev Microbiol 56: 187-209. https://doi.org/10.1146/annurev.micro.56.012302.160705 ![]() |
[60] |
Limoli DH, Jones CJ, Wozniak DJ (2015) Bacterial extracellular polysaccharides in biofilm formation and function. Microbiol Spectrum 3: 10. https://doi.org/10.1128/microbiolspec.MB-0011-2014 ![]() |
[61] |
Magana M, Sereti C, Ioannidis A, et al. (2018) Options and limitations in clinical investigation of bacterial biofilms. Clin Microbiol Rev 31: e00084-16. https://doi.org/10.1128/CMR.00084-16 ![]() |
[62] |
Denef VJ, Mueller RS, Banfield JF (2010) AMD biofilms: using model communities to study microbial evolution and ecological complexity in nature. ISME J 4: 599-610. https://doi.org/10.1038/ismej.2009.158 ![]() |
[63] |
Martinez-Gil M, Goh KGK, Rackaityte E, et al. (2017) YeeJ is an inverse autotransporter from Escherichia coli that binds to peptidoglycan and promotes biofilm formation. Sci Rep 7: 11326. https://doi.org/10.1038/s41598-017-10902-0 ![]() |
[64] | Beloin C, Roux A, Ghigo JM (2008) Escherichia coli biofilms. Curr Top Microbio Immunol 322: 249-289. https://doi.org/10.1007/978-3-540-75418-3_12 |
[65] |
Klemm P, Hjerrild L, Gjermansen M, et al. (2004) Structure-function analysis of the self-recognizing Antigen 43 autotransporter protein from Escherichia coli. Mol Microbiol 51: 283-296. https://doi.org/10.1046/j.1365-2958.2003.03833.x ![]() |
[66] |
Müller D, Benz I, Tapadar D, et al. (2005) Arrangement of the translocator of the autotransporter adhesin involved in diffuse adherence on the bacterial surface. Infect Immun 73: 3851-3859. https://doi.org/10.1128/IAI.73.7.3851-3859.2005 ![]() |
[67] |
Barnhart MM, Chapman MR (2006) Curli biogenesis and function. Annu Rev Microbiol 60: 131-147. https://doi.org/10.1146/annurev.micro.60.080805.142106 ![]() |
[68] |
Tursi SA, Tükel Ç (2018) Curli-Containing enteric biofilms inside and out: Matrix composition, immune recognition, and disease implications. Microbiol Mol Biol Rev 82: e00028-18. https://doi.org/10.1128/MMBR.00028-18 ![]() |
[69] |
Hobley L, Harkins C, MacPhee CE, et al. (2015) Giving structure to the biofilm matrix: an overview of individual strategies and emerging common themes. FEMS Microbiol Rev 39: 649-669. https://doi.org/10.1093/femsre/fuv015 ![]() |
[70] |
Chen XD, Zhang CK, Zhou Z, et al. (2017) Stabilizing effects of bacterial biofilms: EPS penetration and redistribution of bed stability down the sediment profile. J Geophys Res: Biogeosci 122: 3113-3125. https://doi.org/10.1002/2017JG004050 ![]() |
[71] |
Ibáñez de Aldecoa AL, Zafra O, González-Pastor JE (2017) Mechanisms and regulation of extracellular DNA release and its biological roles in microbial communities. Front Microbiol 8: 1390-1390. https://doi.org/10.3389/fmicb.2017.01390 ![]() |
[72] |
Jamal M, Ahmad W, Andleeb S, et al. (2018) Bacterial biofilm and associated infections. J Chin Med Assoc 81: 7-11. https://doi.org/10.1016/j.jcma.2017.07.012 ![]() |
[73] |
Toyofuku M, Inaba T, Kiyokawa T, et al. (2016) Environmental factors that shape biofilm formation. Biosci Biotechnol Biochem 80: 7-12. https://doi.org/10.1080/09168451.2015.1058701 ![]() |
[74] |
Vu B, Chen M, Crawford RJ, et al. (2009) Bacterial extracellular polysaccharides involved in biofilm formation. Molecules (Basel,Switz) 14: 2535-2554. https://doi.org/10.3390/molecules14072535 ![]() |
[75] |
Skariyachan S, Sridhar VS, Packirisamy S, et al. (2018) Recent perspectives on the molecular basis of biofilm formation by Pseudomonas aeruginosa and approaches for treatment and biofilm dispersal. Folia Microbiol 63: 413-432. https://doi.org/10.1007/s12223-018-0585-4 ![]() |
[76] |
Mann EE, Wozniak DJ (2012) Pseudomonas biofilm matrix composition and niche biology. FEMS Microbiol Rev 36: 893-916. https://doi.org/10.1111/j.1574-6976.2011.00322.x ![]() |
[77] |
Zambrano MM, Kolter R (2005) Mycobacterial biofilms: A greasy way to hold it together. Cell 123: 762-764. https://doi.org/10.1016/j.cell.2005.11.011 ![]() |
[78] |
Li YH, Tian X (2012) Quorum sensing and bacterial social interactions in biofilms. Sensors (Basel,Switz) 12: 2519-2538. https://doi.org/10.3390/s120302519 ![]() |
[79] |
Sharma D, Misba L, Khan AU (2019) Antibiotics versus biofilm: an emerging battleground in microbial communities. Antimicrob Resist Infect Control 8: 76. https://doi.org/10.1186/s13756-019-0533-3 ![]() |
[80] |
Shrout JD, Chopp DL, Just CL, et al. (2006) The impact of quorum sensing and swarming motility on Pseudomonas aeruginosa biofilm formation is nutritionally conditional. Mol Microbiol 62: 1264-1277. https://doi.org/10.1111/j.1365-2958.2006.05421.x ![]() |
[81] |
Saygin H, Baysal A (2020) Biofilm formation of clinically important bacteria on bio-based and conventional micro/submicron-sized plastics. Bull Environ Contam Toxicol 105: 18-25. https://doi.org/10.1007/s00128-020-02876-z ![]() |
[82] |
Fulaz S, Vitale S, Quinn L, et al. (2019) Nanoparticle-Biofilm interactions: The role of the EPS matrix. Trends Microbiol 27: 915-926. https://doi.org/10.1016/j.tim.2019.07.004 ![]() |
[83] |
Chakraborty P, Kumar A (2019) The extracellular matrix of mycobacterial biofilms: could we shorten the treatment of mycobacterial infections?. Microb Cell (Graz,Austria) 6: 105-122. https://doi.org/10.15698/mic2019.02.667 ![]() |
[84] |
Yawata Y, Uchiyama H, Nomura N (2010) Visualizing the effects of biofilm structures on the influx of fluorescent material using combined confocal reflection and fluorescent microscopy. Microbes Environ 25: 49-52. https://doi.org/10.1264/jsme2.me09169 ![]() |
[85] |
Holman HY, Miles R, Hao Z, et al. (2009) Real-time chemical imaging of bacterial activity in biofilms using open-channel microfluidics and synchrotron FTIR spectromicroscopy. Anal Chem 81: 8564-8570. https://doi.org/10.1021/ac9015424 ![]() |
[86] |
Mattana S, Alunni Cardinali M, Caponi S, et al. (2017) High-contrast brillouin and raman micro-spectroscopy for simultaneous mechanical and chemical investigation of microbial biofilms. Biophys Chem 229: 123-129. https://doi.org/10.1016/j.bpc.2017.06.008 ![]() |
[87] |
Abadian PN, Tandogan N, Jamieson JJ, et al. (2014) Using surface plasmon resonance imaging to study bacterial biofilms. Biomicrofluidics 8: 021804. https://doi.org/10.1063/1.4867739 ![]() |
[88] |
McGoverin C, Vanholsbeeck F, Dawes JM, et al. (2020) Asia-pacific optical sensors conference: focus issue introduction. Opt Express 28: 21745-21748. https://doi.org/10.1364/OE.401277 ![]() |
[89] |
Yuan Y, Guo T, Qiu X, et al. (2016) Electrochemical surface plasmon resonance fiber-optic sensor: In situ detection of electroactive biofilms. Anal Chem 88: 7609-7616. https://doi.org/10.1021/acs.analchem.6b01314 ![]() |
[90] |
Subramanian S, Huiszoon RC, Chu S, et al. (2019) Microsystems for biofilm characterization and sensing-A review. Biofilm 2: 100015. https://doi.org/10.1016/j.bioflm.2019.100015 ![]() |
[91] |
Stöckl M, Schlegel C, Sydow A, et al. (2016) Membrane separated flow cell for parallelized electrochemical impedance spectroscopy and confocal laser scanning microscopy to characterize electro-active microorganisms. Electrochim Acta 220: 444-452. https://doi.org/10.1016/j.electacta.2016.10.057 ![]() |
[92] |
Estrada-Leypon O, Moya A, Guimera A, et al. (2015) Simultaneous monitoring of Staphylococcus aureus growth in a multi-parametric microfluidic platform using microscopy and impedance spectroscopy. Bioelectrochemistry 105: 56-64. https://doi.org/10.1016/j.bioelechem.2015.05.006 ![]() |
[93] |
Huiszoon RC, Subramanian S, Ramiah Rajasekaran P, et al. (2019) Flexible platform for in situ impedimetric detection and bioelectric effect treatment of Escherichia coli biofilms. IEEE Trans Biomed Eng 66: 1337-1345. https://doi.org/10.1109/tbme.2018.2872896 ![]() |
[94] |
Bayoudh S, Othmane A, Ponsonnet L, et al. (2008) Electrical detection and characterization of bacterial adhesion using electrochemical impedance spectroscopy-based flow chamber. Colloids Surf, A 318: 291-300. https://doi.org/10.1016/j.colsurfa.2008.01.005 ![]() |
[95] |
Bellin DL, Sakhtah H, Zhang Y, et al. (2016) Electrochemical camera chip for simultaneous imaging of multiple metabolites in biofilms. Nat Commun 7: 10535. https://doi.org/10.1038/ncomms10535 ![]() |
[96] |
Marcus IM, Herzberg M, Walker SL, et al. (2012) Pseudomonas aeruginosa attachment on QCM-D sensors: the role of cell and surface hydrophobicities. Langmuir 28: 6396-6402. https://doi.org/10.1021/la300333c ![]() |
[97] |
Olsson AL, van der Mei HC, Busscher HJ, et al. (2009) Influence of cell surface appendages on the bacterium-substratum interface measured real-time using QCM-D. Langmuir 25: 1627-1632. https://doi.org/10.1021/la803301q ![]() |
[98] |
Piasecki T, Guła G, Markwitz P, et al. (2016) Autonomous system for in situ assay of antibiotic activity on bacterial biofilms using viscosity and density sensing quartz tuning forks. Procedia Eng 168: 745-748. https://doi.org/10.1016/j.proeng.2016.11.267 ![]() |
[99] |
Sfaelou S, Karapanagioti HK, Vakros J (2015) Studying the formation of biofilms on supports with different polarity and their efficiency to treat wastewater. J Chem 2015: 734384. https://doi.org/10.1155/2015/734384 ![]() |
[100] |
Khatoon Z, McTiernan CD, Suuronen EJ, et al. (2018) Bacterial biofilm formation on implantable devices and approaches to its treatment and prevention. Heliyon 4: e01067. https://doi.org/10.1016/j.heliyon.2018.e01067 ![]() |
[101] | Revdiwala S, Rajdev BM, Mulla S (2012) Characterization of bacterial etiologic agents of biofilm formation in medical devices in critical care setup. Crit Care Res Pract 2012: 945805. https://doi.org/10.1155/2012/945805 |
[102] |
Trautner BW, Darouiche RO (2004) Role of biofilm in catheter-associated urinary tract infection. Am J Infect Control 32: 177-183. https://doi.org/10.1016/j.ajic.2003.08.005 ![]() |
[103] |
Liu S, Gunawan C, Barraud N, et al. (2016) Understanding, monitoring, and controlling biofilm growth in drinking water distribution systems. Environ Sci Technol 50: 8954-8976. https://doi.org/10.1021/acs.est.6b00835 ![]() |
[104] |
Maes S, Vackier T, Nguyen Huu S, et al. (2019) Occurrence and characterisation of biofilms in drinking water systems of broiler houses. BMC Microbiol 19: 77. https://doi.org/10.1186/s12866-019-1451-5 ![]() |
[105] |
Muhammad MH, Idris AL, Fan X, et al. (2020) Beyond risk: bacterial biofilms and their regulating approaches. Front Microbiol 11: 928-928. https://doi.org/10.3389/fmicb.2020.00928 ![]() |
[106] |
Ashbolt NJ (2015) Microbial contamination of drinking water and human health from community water systems. Curr Environ Health Rep 2: 95-106. https://doi.org/10.1007/s40572-014-0037-5 ![]() |
[107] |
Archer NK, Mazaitis MJ, Costerton JW, et al. (2011) Staphylococcus aureus biofilms: properties, regulation, and roles in human disease. Virulence 2: 445-459. https://doi.org/10.4161/viru.2.5.17724 ![]() |
[108] |
Arciola CR, Campoccia D, Montanaro L (2018) Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 16: 397-409. https://doi.org/10.1038/s41579-018-0019-y ![]() |
[109] |
Boisvert AA, Cheng MP, Sheppard DC, et al. (2016) Microbial biofilms in pulmonary and critical care diseases. Ann Am Thorac Soc 13: 1615-1623. https://doi.org/10.1513/AnnalsATS.201603-194FR ![]() |
[110] |
Gnanadhas DP, Elango M, Datey A, et al. (2015) Chronic lung infection by Pseudomonas aeruginosa biofilm is cured by L-Methionine in combination with antibiotic therapy. Sci Rep 5: 16043. https://doi.org/10.1038/srep16043 ![]() |
[111] |
Minasyan H (2019) Sepsis: mechanisms of bacterial injury to the patient. Scand J Trauma Resusc Emerg Med 27: 19. https://doi.org/10.1186/s13049-019-0596-4 ![]() |
[112] |
VanEpps JS, Younger JG (2016) Implantable device-related infection. Shock (Augusta,Ga) 46: 597-608. https://doi.org/10.1097/SHK.0000000000000692 ![]() |
[113] |
Stewart PS (2002) Mechanisms of antibiotic resistance in bacterial biofilms. Int J Med Microbiol 292: 107-113. https://doi.org/10.1078/1438-4221-00196 ![]() |
[114] |
Vestby LK, Grønseth T, Simm R, et al. (2020) Bacterial biofilm and its role in the pathogenesis of disease. Antibiotics (Basel,Switz) 9: 59. https://doi.org/10.3390/antibiotics9020059 ![]() |
[115] |
González JF, Hahn MM, Gunn JS (2018) Chronic biofilm-based infections: skewing of the immune response. Pathog Dis 76: fty023. https://doi.org/10.1093/femspd/fty023 ![]() |
[116] |
Elias S, Banin E (2012) Multi-species biofilms: living with friendly neighbors. FEMS Microbiol Rev 36: 990-1004. https://doi.org/10.1111/j.1574-6976.2012.00325.x ![]() |
[117] | Hu X, Kang F, Yang B, et al. (2019) Extracellular polymeric substances acting as a permeable barrier hinder the lateral transfer of antibiotic resistance genes. Front Microbiol 10. https://doi.org/10.3389/fmicb.2019.00736 |
[118] |
Singh S, Singh SK, Chowdhury I, et al. (2017) Understanding the mechanism of bacterial biofilms resistance to antimicrobial agents. Open Microbiol J 11: 53-62. https://doi.org/10.2174/1874285801711010053 ![]() |
[119] |
Roy R, Tiwari M, Donelli G, et al. (2018) Strategies for combating bacterial biofilms: A focus on anti-biofilm agents and their mechanisms of action. Virulence 9: 522-554. https://doi.org/10.1080/21505594.2017.1313372 ![]() |
[120] |
Koo H, Allan RN, Howlin RP, et al. (2017) Targeting microbial biofilms: current and prospective therapeutic strategies. Nat Rev Microbiol 15: 740-755. https://doi.org/10.1038/nrmicro.2017.99 ![]() |
[121] |
Igiri BE, Okoduwa SIR, Idoko GO, et al. (2018) Toxicity and bioremediation of heavy metals contaminated ecosystem from tannery wastewater: A review. J Toxicol 2018: 2568038. https://doi.org/10.1155/2018/2568038 ![]() |
[122] |
Singh R, Paul D, Jain RK (2006) Biofilms: implications in bioremediation. Trends Microbiol 14: 389-397. https://doi.org/10.1016/j.tim.2006.07.001 ![]() |
[123] | Bispo PJM, Haas W, Gilmore MS (2015) Biofilms in infections of the eye. Pathogens (Basel,Switz) 4: 111-136. https://doi.org/10.3390/pathogens4010111 |
[124] | Nickel JC, Costerton JW (1992) Bacterial biofilms and catheters: A key to understanding bacterial strategies in catheter-associated urinary tract infection. Can J Infect Dis 3: 261-267. https://doi.org/10.1155/1992/517456 |
[125] | Guynn JB, Poretz DM, Duma RJ (1973) Growth of various bacteria in a variety of intravenous fluids. Am J Hosp Pharm 30: 321-325. https://doi.org/10.1093/ajhp/30.4.321 |
[126] |
Ferrières L, Hancock V, Klemm P (2007) Specific selection for virulent urinary tract infectious Escherichia coli strains during catheter-associated biofilm formation. FEMS Immunol Med Microbiol 51: 212-219. https://doi.org/10.1111/j.1574-695X.2007.00296.x ![]() |
[127] | Delcaru C, Alexandru I, Podgoreanu P, et al. (2016) Microbial biofilms in urinary tract infections and prostatitis: Etiology, pathogenicity, and combating strategies. Pathogens (Basel,Switz) 5: 65. https://doi.org/10.3390/pathogens5040065 |
[128] |
Wiley L, Bridge DR, Wiley LA, et al. (2012) Bacterial biofilm diversity in contact lens-related disease: Emerging role of achromobacter, stenotrophomonas, and delftia. Invest Ophthalmol Visual Sci 53: 3896-3905. https://doi.org/10.1167/iovs.11-8762 ![]() |
[129] |
Robertson DM, Parks QM, Young RL, et al. (2011) Disruption of contact lens-associated Pseudomonas aeruginosa biofilms formed in the presence of neutrophils. Invest Ophthalmol Visual Sci 52: 2844-2850. https://doi.org/10.1167/iovs.10-6469 ![]() |
[130] |
Litzler PY, Benard L, Barbier-Frebourg N, et al. (2007) Biofilm formation on pyrolytic carbon heart valves: influence of surface free energy, roughness, and bacterial species. J Thorac Cardiovasc Surg 134: 1025-1032. https://doi.org/10.1016/j.jtcvs.2007.06.013 ![]() |
[131] | Piper C, Körfer R, Horstkotte D (2001) Prosthetic valve endocarditis. Heart 85: 590. https://doi.org/10.1136/heart.85.5.590 |
[132] |
Schaudinn C, Gorur A, Keller D, et al. (2009) Periodontitis: an archetypical biofilm disease. J Am Dent Assoc 140: 978-986. https://doi.org/10.14219/jada.archive.2009.0307 ![]() |
[133] |
Naginyte M, Do T, Meade J, et al. (2019) Enrichment of periodontal pathogens from the biofilms of healthy adults. Sci Rep 9: 5491. https://doi.org/10.1038/s41598-019-41882-y ![]() |
[134] |
Chandki R, Banthia P, Banthia R (2011) Biofilms: A microbial home. J Indian Soc Periodontol 15: 111-114. https://doi.org/10.4103/0972-124X.84377 ![]() |
[135] |
Brady RA, Leid JG, Calhoun JH, et al. (2008) Osteomyelitis and the role of biofilms in chronic infection. FEMS Immunol Med Microbiol 52: 13-22. https://doi.org/10.1111/j.1574-695X.2007.00357.x ![]() |
[136] |
Masters EA, Trombetta RP, de Mesy Bentley KL, et al. (2019) Evolving concepts in bone infection: redefining “biofilm”, “acute vs. chronic osteomyelitis”, “the immune proteome” and “local antibiotic therapy”. Bone Res 7: 20. https://doi.org/10.1038/s41413-019-0061-z ![]() |
[137] |
Rochford ETJ, Sabaté Brescó M, Zeiter S, et al. (2016) Monitoring immune responses in a mouse model of fracture fixation with and without Staphylococcus aureus osteomyelitis. Bone 83: 82-92. https://doi.org/10.1016/j.bone.2015.10.014 ![]() |
[138] |
Oates A, Bowling FL, Boulton AJM, et al. (2014) The visualization of biofilms in chronic diabetic foot wounds using routine diagnostic microscopy methods. J Diabetes Res 2014: 153586. https://doi.org/10.1155/2014/153586 ![]() |
[139] |
Clinton A, Carter T (2015) Chronic wound biofilms: Pathogenesis and potential therapies. Lab Med 46: 277-284. https://doi.org/10.1309/lmbnswkui4jpn7so ![]() |
[140] |
Bjarnsholt T (2013) The role of bacterial biofilms in chronic infections. APMIS Suppl : 1-51. https://doi.org/10.1111/apm.12099 ![]() |
[141] |
Burmølle M, Thomsen TR, Fazli M, et al. (2010) Biofilms in chronic infections-a matter of opportunity-monospecies biofilms in multispecies infections. FEMS Immunol Med Microbiol 59: 324-336. https://doi.org/10.1111/j.1574-695X.2010.00714.x ![]() |
[142] |
Percival SL, McCarty SM, Lipsky B (2015) Biofilms and wounds: An overview of the evidence. Adv Wound Care 4: 373-381. https://doi.org/10.1089/wound.2014.0557 ![]() |
[143] |
Abdel-Nour M, Duncan C, Low DE, et al. (2013) Biofilms: the stronghold of Legionella pneumophila. Int J Mol Sci 14: 21660-21675. https://doi.org/10.3390/ijms141121660 ![]() |
[144] |
Høiby N, Ciofu O, Bjarnsholt T (2010) Pseudomonas aeruginosa biofilms in cystic fibrosis. Future Microbiol 5: 1663-1674. https://doi.org/10.2217/fmb.10.125 ![]() |
[145] |
Moreau-Marquis S, Stanton BA, O'Toole GA (2008) Pseudomonas aeruginosa biofilm formation in the cystic fibrosis airway. Pulm Pharmacol Ther 21: 595-599. https://doi.org/10.1016/j.pupt.2007.12.001 ![]() |
[146] |
Lalani T, Kanafani ZA, Chu VH, et al. (2006) Prosthetic valve endocarditis due to coagulase-negative staphylococci: findings from the international collaboration on endocarditis merged database. Eur J Clin Microbiol Infect Dis 25: 365-368. https://doi.org/10.1007/s10096-006-0141-z ![]() |
[147] |
van Steenbergen TJM, van Winkelhoff AJ, de Graaff J (1984) Pathogenic synergy: mixed infections in the oral cavity. Antonie van Leeuwenhoek 50: 789-798. https://doi.org/10.1007/BF02386241 ![]() |
[148] |
Chen L, Deng H, Cui H, et al. (2017) Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 9: 7204-7218. https://doi.org/10.18632/oncotarget.23208 ![]() |
[149] |
Kany S, Vollrath JT, Relja B (2019) Cytokines in inflammatory disease. Int J Mol Sci 20: 6008. https://doi.org/10.3390/ijms20236008 ![]() |
[150] |
Hall-Stoodley L, Stoodley P, Kathju S, et al. (2012) Towards diagnostic guidelines for biofilm-associated infections. FEMS Immunol Med Microbiol 65: 127-145. https://doi.org/10.1111/j.1574-695X.2012.00968.x ![]() |
[151] | Olson ME, Ceri H, Morck DW, et al. (2002) Biofilm bacteria: formation and comparative susceptibility to antibiotics. Can J Vet Res 66: 86-92. |
[152] |
Mosaddad SA, Tahmasebi E, Yazdanian A, et al. (2019) Oral microbial biofilms: an update. Eur J Clin Microbiol Infect Dis 38: 2005-2019. https://doi.org/10.1007/s10096-019-03641-9 ![]() |
[153] | Fair RJ, Tor Y (2014) Antibiotics and bacterial resistance in the 21st century. Perspect Med Chem 6: 25-64. https://doi.org/10.4137/PMC.S14459 |
[154] |
Hedberg M, Nord CE (1996) Beta-lactam resistance in anaerobic bacteria: a review. J Chemother 8: 3-16. https://doi.org/10.1179/joc.1996.8.1.3 ![]() |
[155] |
Macià MD, Rojo-Molinero E, Oliver A (2014) Antimicrobial susceptibility testing in biofilm-growing bacteria. Clin Microbiol Infect 20: 981-990. https://doi.org/10.1111/1469-0691.12651 ![]() |
[156] |
Lewis K (2001) Riddle of biofilm resistance. Antimicrob Agents Chemother 45: 999. https://doi.org/10.1128/AAC.45.4.999-1007.2001 ![]() |
[157] |
Kaplan JB, Mlynek KD, Hettiarachchi H, et al. (2018) Extracellular polymeric substance (EPS)-degrading enzymes reduce Staphylococcal surface attachment and biocide resistance on pig skin in vivo. PLoS One 13: e0205526. https://doi.org/10.1371/journal.pone.0205526 ![]() |
[158] |
Powell LC, Pritchard MF, Ferguson EL, et al. (2018) Targeted disruption of the extracellular polymeric network of Pseudomonas aeruginosa biofilms by alginate oligosaccharides. npj Biofilms Microbiomes 4: 13. https://doi.org/10.1038/s41522-018-0056-3 ![]() |
[159] |
Wozniak DJ, Wyckoff TJ, Starkey M, et al. (2003) Alginate is not a significant component of the extracellular polysaccharide matrix of PA14 and PAO1 Pseudomonas aeruginosa biofilms. Proc Natl Acad Sci U S A 100: 7907-7912. https://doi.org/10.1073/pnas.1231792100 ![]() |
[160] |
Madsen JS, Burmølle M, Hansen LH, et al. (2012) The interconnection between biofilm formation and horizontal gene transfer. FEMS Immunol Med Microbiol 65: 183-195. https://doi.org/10.1111/j.1574-695X.2012.00960.x ![]() |
[161] |
Lister PD, Wolter DJ, Hanson ND (2009) Antibacterial-resistant Pseudomonas aeruginosa: clinical impact and complex regulation of chromosomally encoded resistance mechanisms. Clin Microbiol Rev 22: 582-610. https://doi.org/10.1128/CMR.00040-09 ![]() |
[162] |
Wilton M, Charron-Mazenod L, Moore R, et al. (2015) Extracellular DNA acidifies biofilms and induces aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob Agents Chemother 60: 544-553. https://doi.org/10.1128/AAC.01650-15 ![]() |
[163] |
Mulcahy H, Charron-Mazenod L, Lewenza S (2008) Extracellular DNA chelates cations and induces antibiotic resistance in Pseudomonas aeruginosa biofilms. PLoS Pathog 4: e1000213. https://doi.org/10.1371/journal.ppat.1000213 ![]() |
[164] |
Heilmann C, Hartleib J, Hussain MS, et al. (2005) The multifunctional Staphylococcus aureus autolysin aaa mediates adherence to immobilized fibrinogen and fibronectin. Infect Immunol 73: 4793-4802. https://doi.org/10.1128/iai.73.8.4793-4802.2005 ![]() |
[165] |
Heilmann C, Hussain M, Peters G, et al. (1997) Evidence for autolysin-mediated primary attachment of Staphylococcus epidermidis to a polystyrene surface. Mol Microbiol 24: 1013-1024. https://doi.org/10.1046/j.1365-2958.1997.4101774.x ![]() |
[166] |
Ch'ng JH, Chong KKL, Lam LN, et al. (2019) Biofilm-associated infection by enterococci. Nat Rev Microbiol 17: 82-94. https://doi.org/10.1038/s41579-018-0107-z ![]() |
[167] |
Dale JL, Nilson JL, Barnes AMT, et al. (2017) Restructuring of Enterococcus faecalis biofilm architecture in response to antibiotic-induced stress. npj Biofilms Microbiomes 3: 15. https://doi.org/10.1038/s41522-017-0023-4 ![]() |
[168] |
Balaban NQ, Helaine S, Lewis K, et al. (2019) Definitions and guidelines for research on antibiotic persistence. Nat Rev Microbiol 17: 441-448. https://doi.org/10.1038/s41579-019-0196-3 ![]() |
[169] |
Schilcher K, Horswill AR (2020) Staphylococcal biofilm development: Structure, regulation, and treatment strategies. Microbiol Mol Biol Rev 84: e00026-19. https://doi.org/doi:10.1128/MMBR.00026-19 ![]() |
[170] |
Waters EM, Rowe SE, O'Gara JP, et al. (2016) Convergence of Staphylococcus aureus persister and biofilm research: Can biofilms be defined as communities of adherent persister cells?. PLoS Pathog 12: e1006012. https://doi.org/10.1371/journal.ppat.1006012 ![]() |
[171] |
Lister JL, Horswill AR (2014) Staphylococcus aureus biofilms: recent developments in biofilm dispersal. Front Cell Infect Microbiol 4. https://doi.org/10.3389/fcimb.2014.00178 ![]() |
[172] |
Vuotto C, Longo F, Balice MP, et al. (2014) Antibiotic resistance related to biofilm formation in Klebsiella pneumoniae. Pathogens 3: 743-758. https://doi.org/10.3390/pathogens3030743 ![]() |
[173] |
Nirwati H, Sinanjung K, Fahrunissa F, et al. (2019) Biofilm formation and antibiotic resistance of Klebsiella pneumoniae isolated from clinical samples in a tertiary care hospital, Klaten, Indonesia. BMC Proc 13: 20. https://doi.org/10.1186/s12919-019-0176-7 ![]() |
[174] |
Ramos-Vivas J, Chapartegui-González I, Fernández-Martínez M, et al. (2019) Biofilm formation by multidrug resistant Enterobacteriaceae strains isolated from solid organ transplant recipients. Sci Rep 9: 8928. https://doi.org/10.1038/s41598-019-45060-y ![]() |
[175] |
Yang CH, Su PW, Moi SH, et al. (2019) Biofilm formation in acinetobacter baumannii: genotype-phenotype correlation. Molecules (Basel,Switz) 24: 1849. https://doi.org/10.3390/molecules24101849 ![]() |
[176] |
Smani Y, McConnell MJ, Pachón J, et al. (2012) Role of fibronectin in the adhesion of Acinetobacter baumannii to host cells. PLoS One 7: e33073. https://doi.org/10.1371/journal.pone.0033073 ![]() |
[177] |
Gaddy JA, Tomaras AP, Actis LA (2009) The Acinetobacter baumannii 19606 OmpA protein plays a role in biofilm formation on abiotic surfaces and in the interaction of this pathogen with eukaryotic cells. Infect Immun 77: 3150-3160. https://doi.org/10.1128/iai.00096-09 ![]() |
[178] |
Dumaru R, Baral R, Shrestha LB (2019) Study of biofilm formation and antibiotic resistance pattern of gram-negative Bacilli among the clinical isolates at BPKIHS, Dharan. BMC Res Notes 12: 38. https://doi.org/10.1186/s13104-019-4084-8 ![]() |
[179] |
Poulsen LK, Ballard G, Stahl DA (1993) Use of rRNA fluorescence in situ hybridization for measuring the activity of single cells in young and established biofilms. Appl Environ Microbiol 59: 1354-1360. https://doi.org/10.1128/aem.59.5.1354-1360.1993 ![]() |
[180] |
Sternberg C, Christensen BB, Johansen T, et al. (1999) Distribution of bacterial growth activity in flow-chamber biofilms. Appl Environ Microbiol 65: 4108-4117. https://doi.org/10.1128/aem.65.9.4108-4117.1999 ![]() |
[181] |
Mitchison JM (1969) Enzyme synthesis in synchronous cultures. Science 165: 657-663. https://doi.org/10.1126/science.165.3894.657 ![]() |
[182] | Gilbert P, Maira-Litran T, McBain AJ, et al. (2002) The physiology and collective recalcitrance of microbial biofilm communities. Adv Microb Physiol 46: 202-256. https://doi.org/10.1016/S0065-2911(02)46005-5 |
[183] |
Walters MC, Roe F, Bugnicourt A, et al. (2003) Contributions of antibiotic penetration, oxygen limitation, and low metabolic activity to tolerance of Pseudomonas aeruginosa biofilms to ciprofloxacin and tobramycin. Antimicrob Agents Chemother 47: 317-323. https://doi.org/10.1128/aac.47.1.317-323.2003 ![]() |
[184] |
Tresse O, Jouenne T, Junter GA (1995) The role of oxygen limitation in the resistance of agar-entrapped, sessile-like Escherichia coli to aminoglycoside and beta-lactam antibiotics. J Antimicrob Chemother 36: 521-526. https://doi.org/10.1093/jac/36.3.521 ![]() |
[185] |
Hall CW, Mah TF (2017) Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol Rev 41: 276-301. https://doi.org/10.1093/femsre/fux010 ![]() |
[186] |
Zheng Z, Stewart PS (2004) Growth limitation of Staphylococcus epidermidis in biofilms contributes to rifampin tolerance. Biofilms 1: 31-35. https://doi.org/10.1017/S1479050503001042 ![]() |
[187] |
Mah TFC, O'Toole GA (2001) Mechanisms of biofilm resistance to antimicrobial agents. Trends Microbiol 9: 34-39. https://doi.org/10.1016/S0966-842X(00)01913-2 ![]() |
[188] |
Zhao X, Yu Z, Ding T (2020) Quorum-Sensing regulation of antimicrobial resistance in bacteria. Microorganisms 8: 425. https://doi.org/10.3390/microorganisms8030425 ![]() |
[189] |
Shih PC, Huang CT (2002) Effects of quorum-sensing deficiency on Pseudomonas aeruginosa biofilm formation and antibiotic resistance. J Antimicrob Chemother 49: 309-314. https://doi.org/10.1093/jac/49.2.309 ![]() |
[190] | Algburi A, Comito N, Kashtanov D, et al. (2017) Control of biofilm formation: Antibiotics and beyond. Appl Environ Microbiol 83: e02508-02516. https://doi.org/10.1128/AEM.02508-16 |
[191] |
Jiao Y, Tay FR, Niu Ln, et al. (2019) Advancing antimicrobial strategies for managing oral biofilm infections. Int J Oral Sci 11: 28. https://doi.org/10.1038/s41368-019-0062-1 ![]() |
[192] |
Klemm P, Vejborg RM, Hancock V (2010) Prevention of bacterial adhesion. Appl Microbiol Biotechnol 88: 451-459. https://doi.org/10.1007/s00253-010-2805-y ![]() |
[193] |
Veerachamy S, Yarlagadda T, Manivasagam G, et al. (2014) Bacterial adherence and biofilm formation on medical implants: A review. Proc Inst Mech Eng, Part H 228: 1083-1099. https://doi.org/10.1177/0954411914556137 ![]() |
[194] |
Chow JY, Yang Y, Tay SB, et al. (2014) Disruption of biofilm formation by the human pathogen Acinetobacter baumannii using engineered quorum-quenching lactonases. Antimicrob Agents Chemother 58: 1802-1805. https://doi.org/10.1128/AAC.02410-13 ![]() |
[195] |
Brown HL, Reuter M, Hanman K, et al. (2015) Prevention of biofilm formation and removal of existing biofilms by extracellular DNases of campylobacter jejuni. PLoS One 10: e0121680. https://doi.org/10.1371/journal.pone.0121680 ![]() |
[196] | Jiang Y, Geng M, Bai L (2020) Targeting biofilms therapy: Current research strategies and development hurdles. Microorganisms 8. https://doi.org/10.3390/microorganisms8081222 |
[197] |
Gil C, Solano C, Burgui S, et al. (2014) Biofilm matrix exoproteins induce a protective immune response against Staphylococcus aureus biofilm infection. Infect Immun 82: 1017-1029. https://doi.org/10.1128/IAI.01419-13 ![]() |
[198] | Jiang Q, Jin Z, Sun B (2018) MgrA negatively regulates biofilm formation and detachment by repressing the expression of psm operons in Staphylococcus aureus. Appl Environ Microbiol 84. https://doi.org/10.1128/aem.01008-18 |
[199] |
Fleming D, Rumbaugh KP (2017) Approaches to dispersing medical biofilms. Microorganisms 5: 15. https://doi.org/10.3390/microorganisms5020015 ![]() |
[200] |
Gallant CV, Daniels C, Leung JM, et al. (2005) Common beta-lactamases inhibit bacterial biofilm formation. Mol Microbiol 58: 1012-1024. https://doi.org/10.1111/j.1365-2958.2005.04892.x ![]() |
[201] |
Reza A, Sutton JM, Rahman KM (2019) Effectiveness of efflux pump inhibitors as biofilm disruptors and resistance breakers in Gram-Negative (ESKAPEE) bacteria. Antibiotics (Basel,Switz) 8: 229. https://doi.org/10.3390/antibiotics8040229 ![]() |
[202] |
Ciofu O, Rojo-Molinero E, Macià MD, et al. (2017) Antibiotic treatment of biofilm infections. APMIS 125: 304-319. https://doi.org/10.1111/apm.12673 ![]() |
[203] |
Brooun A, Liu S, Lewis K (2000) A dose-response study of antibiotic resistance in Pseudomonas aeruginosa biofilms. Antimicrob Agents Chemother 44: 640-646. https://doi.org/10.1128/aac.44.3.640-646.2000 ![]() |
[204] |
Danese PN (2002) Antibiofilm Approaches: Prevention of Catheter Colonization. Chem Biol 9: 873-880. https://doi.org/10.1016/S1074-5521(02)00192-8 ![]() |
[205] |
Flemming HC, Ridgway H (2009) Biofilm control: Conventional and alternative approaches. Marine and Industrial Biofouling. Heidelberg: Springer Berlin Heidelberg 103-117. https://doi.org/10.1007/978-3-540-69796-1_5 ![]() |
[206] |
Sambanthamoorthy K, Gokhale AA, Lao W, et al. (2011) Identification of a novel benzimidazole that inhibits bacterial biofilm formation in a broad-spectrum manner. Antimicrob Agents Chemother 55: 4369. https://doi.org/10.1128/AAC.00583-11 ![]() |
[207] | Dinicola S, De Grazia S, Carlomagno G, et al. (2014) N-acetylcysteine as powerful molecule to destroy bacterial biofilms. A systematic review. Eur Rev Med Pharmacol Sci 18: 2942-2948. Available from: https://europepmc.org/article/MED/25339490 |
[208] |
Abraham NM, Lamlertthon S, Fowler VG, et al. (2012) Chelating agents exert distinct effects on biofilm formation in Staphylococcus aureus depending on strain background: role for clumping factor B. J Med Microbiol 61: 1062-1070. https://doi.org/10.1099/jmm.0.040758-0 ![]() |
[209] |
Antoci V, Adams CS, Parvizi J, et al. (2008) The inhibition of Staphylococcus epidermidis biofilm formation by vancomycin-modified titanium alloy and implications for the treatment of periprosthetic infection. Biomaterials 29: 4684-4690. https://doi.org/10.1016/j.biomaterials.2008.08.016 ![]() |
[210] |
Ma Y, Chen M, Jones JE, et al. (2012) Inhibition of Staphylococcus epidermidis biofilm by trimethylsilane plasma coating. Antimicrob Agents Chemother 56: 5923-5937. https://doi.org/10.1128/aac.01739-12 ![]() |
[211] |
Sanyasi S, Majhi RK, Kumar S, et al. (2016) Polysaccharide-capped silver nanoparticles inhibit biofilm formation and eliminate multi-drug-resistant bacteria by disrupting bacterial cytoskeleton with reduced cytotoxicity towards mammalian cells. Sci Rep 6: 24929. https://doi.org/10.1038/srep24929 ![]() |
[212] |
AshaRani PV, Low Kah Mun G, Hande MP, et al. (2009) Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano 3: 279-290. https://doi.org/10.1021/nn800596w ![]() |
[213] |
Raie DS, Mhatre E, Thiele M, et al. (2017) Application of quercetin and its bio-inspired nanoparticles as anti-adhesive agents against Bacillus subtilis attachment to surface. Mater Sci Eng, C 70: 753-762. https://doi.org/10.1016/j.msec.2016.09.038 ![]() |
[214] |
Hume EB, Baveja J, Muir B, et al. (2004) The control of Staphylococcus epidermidis biofilm formation and in vivo infection rates by covalently bound furanones. Biomaterials 25: 5023-5030. https://doi.org/10.1016/j.biomaterials.2004.01.048 ![]() |
[215] |
Li H, Bao H, Bok KX, et al. (2016) High durability and low toxicity antimicrobial coatings fabricated by quaternary ammonium silane copolymers. Biomater Sci 4: 299-309. https://doi.org/10.1039/C5BM00353A ![]() |
[216] |
Trentin DS, Silva DB, Frasson AP, et al. (2015) Natural green coating inhibits adhesion of clinically important bacteria. Sci Rep 5: 8287. https://doi.org/10.1038/srep08287 ![]() |
[217] |
Chen M, Yu Q, Sun H (2013) Novel strategies for the prevention and treatment of biofilm related infections. Int J Mol Sci 14: 18488-18501. https://doi.org/10.3390/ijms140918488 ![]() |
[218] |
Tran PL, Lowry N, Campbell T, et al. (2012) An organoselenium compound inhibits Staphylococcus aureus biofilms on hemodialysis catheters in vivo. Antimicrob Agents Chemother 56: 972-978. https://doi.org/10.1128/aac.05680-11 ![]() |
[219] |
Zhao X, Zhao F, Wang J, et al. (2017) Biofilm formation and control strategies of foodborne pathogens: food safety perspectives. RSC Adv 7: 36670-36683. https://doi.org/10.1039/C7RA02497E ![]() |
[220] |
de Sousa DG, Harvey LA, Dorsch S, et al. (2018) Interventions involving repetitive practice improve strength after stroke: a systematic review. J Physiother 64: 210-221. https://doi.org/10.1016/j.jphys.2018.08.004 ![]() |
[221] |
Chappell TC, Nair NU (2020) Engineered lactobacilli display anti-biofilm and growth suppressing activities against Pseudomonas aeruginosa. npj Biofilms Microbiomes 6: 48. https://doi.org/10.1038/s41522-020-00156-6 ![]() |
[222] |
Lu L, Hu W, Tian Z, et al. (2019) Developing natural products as potential anti-biofilm agents. Chin Med 14: 11. https://doi.org/10.1186/s13020-019-0232-2 ![]() |
[223] |
Bendaoud M, Vinogradov E, Balashova NV, et al. (2011) Broad-spectrum biofilm inhibition by Kingella kingae exopolysaccharide. J Bacteriol Res 193: 3879-3886. https://doi.org/10.1128/JB.00311-11 ![]() |
[224] |
Mi L, Licina GA, Jiang S (2014) Nonantibiotic-Based Pseudomonas aeruginosa biofilm inhibition with osmoprotectant analogues. ACS Sustainable Chem Eng 2: 2448-2453. https://doi.org/10.1021/sc500468a ![]() |
[225] |
Kuang X, Chen V, Xu X (2018) Novel approaches to the control of oral microbial biofilms. BioMed Res Int 2018: 6498932. https://doi.org/10.1155/2018/6498932 ![]() |
[226] |
Chen H, Zhang B, Weir MD, et al. (2020) S. mutans gene-modification and antibacterial resin composite as dual strategy to suppress biofilm acid production and inhibit caries. J Dent 93: 103278. https://doi.org/10.1016/j.jdent.2020.103278 ![]() |
[227] |
Pires DP, Melo LDR, Vilas Boas D, et al. (2017) Phage therapy as an alternative or complementary strategy to prevent and control biofilm-related infections. Curr Opin Microbiol 39: 48-56. https://doi.org/10.1016/j.mib.2017.09.004 ![]() |
[228] |
Dickey J, Perrot V (2019) Adjunct phage treatment enhances the effectiveness of low antibiotic concentration against Staphylococcus aureus biofilms in vitro. PLoS One 14: e0209390. https://doi.org/10.1371/journal.pone.0209390 ![]() |
[229] |
Pires DP, Vilas Boas D, Sillankorva S, et al. (2015) Phage therapy: a step forward in the treatment of Pseudomonas aeruginosa infections. J Virol 89: 7449-7456. https://doi.org/10.1128/jvi.00385-15 ![]() |
[230] |
Fu W, Forster T, Mayer O, et al. (2010) Bacteriophage cocktail for the prevention of biofilm formation by Pseudomonas aeruginosa on catheters in an in vitro model system. Antimicrob Agents Chemother 54: 397-404. https://doi.org/10.1128/aac.00669-09 ![]() |
[231] |
Paluch E, Rewak-Soroczyńska J, Jędrusik I, et al. (2020) Prevention of biofilm formation by quorum quenching. Appl Microbiol Biotechnol 104: 1871-1881. https://doi.org/10.1007/s00253-020-10349-w ![]() |
[232] | Singh VK, Mishra A, Jha B (2017) Anti-quorum sensing and anti-biofilm activity of delftia tsuruhatensis extract by attenuating the quorum sensing-controlled virulence factor production in Pseudomonas aeruginosa. Front Cell Infect Microbiol 7. https://doi.org/10.3389/fcimb.2017.00337 |
[233] |
Brackman G, Defoirdt T, Miyamoto C, et al. (2008) Cinnamaldehyde and cinnamaldehyde derivatives reduce virulence in Vibrio spp. by decreasing the DNA-binding activity of the quorum sensing response regulator LuxR. BMC Microbiol 8: 149. https://doi.org/10.1186/1471-2180-8-149 ![]() |
[234] |
Kalaiarasan E, Thirumalaswamy K, Harish BN, et al. (2017) Inhibition of quorum sensing-controlled biofilm formation in Pseudomonas aeruginosa by quorum-sensing inhibitors. Microb Pathog 111: 99-107. https://doi.org/10.1016/j.micpath.2017.08.017 ![]() |
[235] | Bulman ZP, Ly NS, Lenhard JR, et al. (2017) Influence of rhlR and lasR on polymyxin pharmacodynamics in Pseudomonas aeruginosa and implications for quorum sensing inhibition with azithromycin. Antimicrob Agents Chemother 61. https://doi.org/10.1128/aac.00096-16 |
[236] |
Maura D, Rahme LG (2017) Pharmacological inhibition of the Pseudomonas aeruginosa MvfR quorum-sensing system interferes with biofilm formation and potentiates antibiotic-mediated biofilm disruption. Antimicrob Agents Chemother 61. https://doi.org/10.1128/aac.01362-17 ![]() |
[237] |
Stenvang M, Dueholm MS, Vad BS, et al. (2016) Epigallocatechin gallate remodels overexpressed functional amyloids in Pseudomonas aeruginosa and increases biofilm susceptibility to antibiotic treatment*. J Biol Chem 291: 26540-26553. https://doi.org/10.1074/jbc.M116.739953 ![]() |
[238] |
Guo Q, Wei Y, Xia B, et al. (2016) Identification of a small molecule that simultaneously suppresses virulence and antibiotic resistance of Pseudomonas aeruginosa. Sci Rep 6: 19141. https://doi.org/10.1038/srep19141 ![]() |
[239] |
Freire MO, Devaraj A, Young A, et al. (2017) A bacterial-biofilm-induced oral osteolytic infection can be successfully treated by immuno-targeting an extracellular nucleoid-associated protein. Mol Oral Microbiol 32: 74-88. https://doi.org/10.1111/omi.12155 ![]() |
[240] | Sun D, Accavitti MA, Bryers JD (2005) Inhibition of biofilm formation by monoclonal antibodies against Staphylococcus epidermidis RP62A accumulation-associated protein. Clin Diagn Lab Immunol 12: 93-100. https://doi.org/10.1128/CDLI.12.1.93-100.2005 |
[241] |
Estellés A, Woischnig AK, Liu K, et al. (2016) A high-affinity native human antibody disrupts biofilm from Staphylococcus aureus bacteria and potentiates antibiotic efficacy in a mouse implant infection model. Antimicrob Agents Chemother 60: 2292-2301. https://doi.org/10.1128/aac.02588-15 ![]() |
[242] |
Hall AE, Domanski PJ, Patel PR, et al. (2003) Characterization of a protective monoclonal antibody recognizing Staphylococcus aureus MSCRAMM protein clumping factor A. Infect Immun 71: 6864-6870. https://doi.org/10.1128/iai.71.12.6864-6870.2003 ![]() |
[243] |
Visai L, Xu Y, Casolini F, et al. (2000) Monoclonal antibodies to CNA, a collagen-binding microbial surface component recognizing adhesive matrix molecules, detach Staphylococcus aureus from a collagen substrate. J Biol Chem 275: 39837-39845. https://doi.org/10.1074/jbc.M005297200 ![]() |
[244] |
Rennermalm A, Li YH, Bohaufs L, et al. (2001) Antibodies against a truncated Staphylococcus aureus fibronectin-binding protein protect against dissemination of infection in the rat. Vaccine 19: 3376-3383. https://doi.org/10.1016/s0264-410x(01)00080-9 ![]() |
[245] |
Belyi Y, Rybolovlev I, Polyakov N, et al. (2018) Staphylococcus aureus surface protein G is an immunodominant protein and a possible target in an anti-biofilm drug development. Open Microbiol J 12: 94-106. https://doi.org/10.2174/1874285801812010094 ![]() |
[246] |
Haghighat S, Siadat SD, Sorkhabadi SM, et al. (2017) Cloning, expression and purification of autolysin from methicillin-resistant Staphylococcus aureus: potency and challenge study in Balb/c mice. Mol Immunol 82: 10-18. https://doi.org/10.1016/j.molimm.2016.12.013 ![]() |
[247] |
Nair N, Vinod V, Suresh MK, et al. (2015) Amidase, a cell wall hydrolase, elicits protective immunity against Staphylococcus aureus and S. epidermidis. Int J Biol Macromol 77: 314-321. https://doi.org/10.1016/j.ijbiomac.2015.03.047 ![]() |
[248] |
Varrone JJ, de Mesy Bentley KL, Bello-Irizarry SN, et al. (2014) Passive immunization with anti-glucosaminidase monoclonal antibodies protects mice from implant-associated osteomyelitis by mediating opsonophagocytosis of Staphylococcus aureus megaclusters. J Orthop Res 32: 1389-1396. https://doi.org/10.1002/jor.22672 ![]() |
[249] |
van den Berg S, Bonarius HP, van Kessel KP, et al. (2015) A human monoclonal antibody targeting the conserved Staphylococcal antigen IsaA protects mice against Staphylococcus aureus bacteremia. Int J Med Microbiol 305: 55-64. https://doi.org/10.1016/j.ijmm.2014.11.002 ![]() |
[250] |
Liu B, Park S, Thompson CD, et al. (2017) Antibodies to Staphylococcus aureus capsular polysaccharides 5 and 8 perform similarly in vitro but are functionally distinct in vivo. Virulence 8: 859-874. https://doi.org/10.1080/21505594.2016.1270494 ![]() |
[251] |
Weisman LE (2007) Antibody for the prevention of neonatal noscocomial Staphylococcal infection: a review of the literature. Arch Pediatr 14 Suppl 1: S31-34. https://doi.org/10.1016/s0929-693x(07)80008-x ![]() |
[252] |
Raafat D, Otto M, Reppschläger K, et al. (2019) Fighting Staphylococcus aureus biofilms with monoclonal antibodies. Trends Microbiol 27: 303-322. https://doi.org/10.1016/j.tim.2018.12.009 ![]() |
[253] |
Freire-Moran L, Aronsson B, Manz C, et al. (2011) Critical shortage of new antibiotics in development against multidrug-resistant bacteria-time to react is now. Drug Resist Updat 14: 118-124. https://doi.org/10.1016/j.drup.2011.02.003 ![]() |
[254] |
Banerjee M, Moulick S, Bhattacharya KK, et al. (2017) Attenuation of Pseudomonas aeruginosa quorum sensing, virulence and biofilm formation by extracts of Andrographis paniculata. Microb Pathog 113: 85-93. https://doi.org/10.1016/j.micpath.2017.10.023 ![]() |
[255] |
Zhang L, Bao M, Liu B, et al. (2020) Effect of andrographolide and its analogs on bacterial infection: A review. Pharmacology 105: 123-134. https://doi.org/10.1159/000503410 ![]() |
[256] |
Rasool U, SP, Parveen A, et al. (2018) Efficacy of andrographis paniculata against extended spectrum β-lactamase (ESBL) producing E. coli. BMC Complementary Altern Med 18: 244-244. https://doi.org/10.1186/s12906-018-2312-8 ![]() |
[257] |
Carrol DH, Chassagne F, Dettweiler M, et al. (2020) Antibacterial activity of plant species used for oral health against Porphyromonas gingivalis. PLoS One 15: e0239316. https://doi.org/10.1371/journal.pone.0239316 ![]() |
[258] |
Gerits E, Verstraeten N, Michiels J (2017) New approaches to combat Porphyromonas gingivalis biofilms. J Oral Microbiol 9: 1300366. https://doi.org/10.1080/20002297.2017.1300366 ![]() |
[259] |
Liu G, Xiang H, Tang X, et al. (2011) Transcriptional and functional analysis shows sodium houttuyfonate-mediated inhibition of autolysis in Staphylococcus aureus. Molecules (Basel,Switz) 16: 8848-8865. https://doi.org/10.3390/molecules16108848 ![]() |
[260] |
Zhou J, Bi S, Chen H, et al. (2017) Anti-Biofilm and antivirulence activities of metabolites from plectosphaerella cucumerina against Pseudomonas aeruginosa. Front Microbiol 8: 769. https://doi.org/10.3389/fmicb.2017.00769 ![]() |
[261] |
Rabin N, Zheng Y, Opoku-Temeng C, et al. (2015) Agents that inhibit bacterial biofilm formation. Future Med Chem 7: 647-671. https://doi.org/10.4155/fmc.15.7 ![]() |
[262] |
Xiang H, Cao F, Ming D, et al. (2017) Aloe-emodin inhibits Staphylococcus aureus biofilms and extracellular protein production at the initial adhesion stage of biofilm development. Appl Microbiol Biotechnol 101: 6671-6681. https://doi.org/10.1007/s00253-017-8403-5 ![]() |
[263] |
Jakobsen TH, van Gennip M, Phipps RK, et al. (2012) Ajoene, a sulfur-rich molecule from garlic, inhibits genes controlled by quorum sensing. Antimicrob Agents Chemother 56: 2314-2325. https://doi.org/10.1128/AAC.05919-11 ![]() |
[264] |
Balcázar JL, Vendrell D, de Blas I, et al. (2008) Characterization of probiotic properties of lactic acid bacteria isolated from intestinal microbiota of fish. Aquaculture 278: 188-191. https://doi.org/10.1016/j.aquaculture.2008.03.014 ![]() |
[265] |
Didinen BI, Onuk EE, Metin S, et al. (2018) Identification and characterization of lactic acid bacteria isolated from rainbow trout (Oncorhynchus mykiss, Walbaum 1792), with inhibitory activity against Vagococcus salmoninarum and Lactococcus garvieae. Aquacult Nutr 24: 400-407. https://doi.org/10.1111/anu.12571 ![]() |
[266] |
Ben Taheur F, Kouidhi B, Fdhila K, et al. (2016) Anti-bacterial and anti-biofilm activity of probiotic bacteria against oral pathogens. Microb Pathog 97: 213-220. https://doi.org/10.1016/j.micpath.2016.06.018 ![]() |
[267] |
Lee DK, Park SY, An HM, et al. (2011) Antimicrobial activity of Bifidobacterium spp. isolated from healthy adult Koreans against cariogenic microflora. Arch Oral Biol 56: 1047-1054. https://doi.org/10.1016/j.archoralbio.2011.03.002 ![]() |
[268] |
Schwendicke F, Horb K, Kneist S, et al. (2014) Effects of heat-inactivated Bifidobacterium BB12 on cariogenicity of Streptococcus mutans in vitro. Arch Oral Biol 59: 1384-1390. https://doi.org/10.1016/j.archoralbio.2014.08.012 ![]() |
[269] |
Suzuki N, Yoneda M, Hatano Y, et al. (2011) Enterococcus faecium WB2000 inhibits biofilm formation by oral cariogenic Streptococci. Int J Dent 2011: 834151. https://doi.org/10.1155/2011/834151 ![]() |
[270] |
Sassone-Corsi M, Raffatellu M (2015) No vacancy: how beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J immunol 194: 4081-4087. https://doi.org/10.4049/jimmunol.1403169 ![]() |
[271] |
Tuomola EM, Ouwehand AC, Salminen SJ (1999) The effect of probiotic bacteria on the adhesion of pathogens to human intestinal mucus. FEMS Immunol Med Microbiol 26: 137-142. https://doi.org/10.1111/j.1574-695X.1999.tb01381.x ![]() |
[272] |
Yan F, Polk DB (2011) Probiotics and immune health. Curr Opin Gastroenterol 27: 496-501. https://doi.org/10.1097/MOG.0b013e32834baa4d ![]() |
[273] |
Hegarty JW, Guinane CM, Ross RP, et al. (2016) Bacteriocin production: a relatively unharnessed probiotic trait?. F1000Res 5: 2587. https://doi.org/10.12688/f1000research.9615.1 ![]() |
[274] |
Corcoran BM, Stanton C, Fitzgerald GF, et al. (2005) Survival of probiotic lactobacilli in acidic environments is enhanced in the presence of metabolizable sugars. Appl Environ Microbiol 71: 3060-3067. https://doi.org/10.1128/AEM.71.6.3060-3067.2005 ![]() |
[275] |
Vuotto C, Longo F, Donelli G (2014) Probiotics to counteract biofilm-associated infections: promising and conflicting data. Int J Oral Sci 6: 189-194. https://doi.org/10.1038/ijos.2014.52 ![]() |
[276] |
Fang K, Jin X, Hong SH (2018) Probiotic Escherichia coli inhibits biofilm formation of pathogenic E. coli via extracellular activity of DegP. Sci Rep 8: 4939. https://doi.org/10.1038/s41598-018-23180-1 ![]() |
[277] |
Jaffar N, Ishikawa Y, Mizuno K, et al. (2016) Mature biofilm degradation by potential probiotics: Aggregatibacter actinomycetemcomitans versus LactoBacillus spp. PLoS One 11: e0159466. https://doi.org/10.1371/journal.pone.0159466 ![]() |
[278] |
Mathur H, Field D, Rea MC, et al. (2018) Fighting biofilms with lantibiotics and other groups of bacteriocins. npj Biofilms Microbiomes 4: 9. https://doi.org/10.1038/s41522-018-0053-6 ![]() |
[279] |
Hertzberger R, Arents J, Dekker Henk L, et al. (2014) H2O2 production in species of the LactoBacillus acidophilus group: a central role for a novel NADH-Dependent flavin reductase. Appl Environ Microbiol 80: 2229-2239. https://doi.org/10.1128/AEM.04272-13 ![]() |
[280] |
Salas-Jara MJ, Ilabaca A, Vega M, et al. (2016) Biofilm forming LactoBacillus: New challenges for the development of probiotics. Microorganisms 4: 35. https://doi.org/10.3390/microorganisms4030035 ![]() |
[281] |
Jones SE, Versalovic J (2009) Probiotic LactoBacillus reuteribiofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol 9: 35. https://doi.org/10.1186/1471-2180-9-35 ![]() |
[282] |
Barzegari A, Kheyrolahzadeh K, Hosseiniyan Khatibi SM, et al. (2020) The battle of probiotics and their derivatives against biofilms. Infect Drug Resist 13: 659-672. https://doi.org/10.2147/IDR.S232982 ![]() |
[283] |
Miquel S, Lagrafeuille R, Souweine B, et al. (2016) Anti-biofilm activity as a health issue. Front Microbiol 7: 592-592. https://doi.org/10.3389/fmicb.2016.00592 ![]() |
[284] |
Tan L, Fu J, Feng F, et al. (2020) Engineered probiotics biofilm enhances osseointegration via immunoregulation and anti-infection. Sci Adv 6: eaba5723. https://doi.org/10.1126/sciadv.aba5723 ![]() |
[285] |
Carvalho FM, Teixeira-Santos R, Mergulhão FJM, et al. (2020) The use of probiotics to fight biofilms in medical devices: A systematic review and meta-analysis. Microorganisms 9. https://doi.org/10.3390/microorganisms9010027 ![]() |
[286] |
Gholizadeh P, Aghazadeh M, Asgharzadeh M, et al. (2017) Suppressing the CRISPR/Cas adaptive immune system in bacterial infections. Eur J Clin Microbiol Infect Dis 36: 2043-2051. https://doi.org/10.1007/s10096-017-3036-2 ![]() |
[287] |
Yao R, Liu D, Jia X, et al. (2018) CRISPR-Cas9/Cas12a biotechnology and application in bacteria. Synth Syst Biotechnol 3: 135-149. https://doi.org/10.1016/j.synbio.2018.09.004 ![]() |
[288] |
Jiang W, Bikard D, Cox D, et al. (2013) RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol 31: 233-239. https://doi.org/10.1038/nbt.2508 ![]() |
[289] |
Goren M, Yosef I, Qimron U (2017) Sensitizing pathogens to antibiotics using the CRISPR-Cas system. Drug Resist Updat 30: 1-6. https://doi.org/10.1016/j.drup.2016.11.001 ![]() |
[290] |
Touchon M, Charpentier S, Pognard D, et al. (2012) Antibiotic resistance plasmids spread among natural isolates of Escherichia coli in spite of CRISPR elements. Microbiology (Reading) 158: 2997-3004. https://doi.org/10.1099/mic.0.060814-0 ![]() |
[291] |
Hale CR, Majumdar S, Elmore J, et al. (2012) Essential features and rational design of CRISPR RNAs that function with the Cas RAMP module complex to cleave RNAs. Mol Cell 45: 292-302. https://doi.org/10.1016/j.molcel.2011.10.023 ![]() |
[292] |
Bikard D, Euler CW, Jiang W, et al. (2014) Exploiting CRISPR-Cas nucleases to produce sequence-specific antimicrobials. Nat Biotechnol 32: 1146-1150. https://doi.org/10.1038/nbt.3043 ![]() |
[293] |
Vercoe RB, Chang JT, Dy RL, et al. (2013) Cytotoxic chromosomal targeting by CRISPR/Cas systems can reshape bacterial genomes and expel or remodel pathogenicity islands. PLoS Genet 9: e1003454. https://doi.org/10.1371/journal.pgen.1003454 ![]() |
[294] |
Gholizadeh P, Köse Ş, Dao S, et al. (2020) How CRISPR-Cas system could be used to combat antimicrobial resistance. Infect Drug Resist 13: 1111-1121. https://doi.org/10.2147/IDR.S247271 ![]() |
[295] |
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. https://doi.org/10.3389/fcimb.2017.00214 ![]() |
[296] |
Gong T, Tang B, Zhou X, et al. (2018) Genome editing in Streptococcus mutans through self-targeting CRISPR arrays. Mol Oral Microbiol 33: 440-449. https://doi.org/10.1111/omi.12247 ![]() |
[297] |
Garrido V, Piñero-Lambea C, Rodriguez-Arce I, et al. (2021) Engineering a genome-reduced bacterium to eliminate Staphylococcus aureus biofilms in vivo. Mol Syst Biol 17: e10145. https://doi.org/10.15252/msb.202010145 ![]() |
1. | Liya Merzon, Sofia Tauriainen, Ana Triana, Tarmo Nurmi, Hanna Huhdanpää, Minna Mannerkoski, Eeva T. Aronen, Mikhail Kantonistov, Linda Henriksson, Emiliano Macaluso, Juha Salmi, Federico Giove, Real-world goal-directed behavior reveals aberrant functional brain connectivity in children with ADHD, 2025, 20, 1932-6203, e0319746, 10.1371/journal.pone.0319746 |
Site | Age | Female/Male | Control | ADHD | Total |
NYU | 7–18 | 76/140 | 98 | 118 | 216 |
PU | 8–17 | 52/142 | 116 | 78 | 194 |
KKI | 8–13 | 37/46 | 61 | 22 | 83 |
NI | 11–22 | 17/31 | 23 | 25 | 48 |
Region name | Region index | Region name | Region index |
Olfactory cortex | 21–22 | Amygdala | 41–42 |
Insula | 29–30 | Caudate | 71–72 |
Anterior cingulate | 31–32 | Putamen | 73–74 |
Middle cingulate | 33–34 | Pallidum | 75–76 |
Posterior cingulate | 35–36 | Thalamus | 77–78 |
Hippocampus | 37–38 | Cerebellum | 91–108 |
Parahippocampal | 39–40 | Vermis | 109–116 |
Layer size* | |
Dense 1 | (25, 10) |
Dense 2 | (10, 25) |
Dense 3 | (10, 8) |
Dense 4 | (8, 8) |
Dense 5 | (8, 2) |
* The parameters (a, b) describe a dense unit with input size of a and output size of b. |
Site | Accuracy (%) | Sensitivity (%) | Specificity (%) | AUROC | AUPRC | MCC | F1 |
Without ensemble learning | |||||||
NYU | 91.94 | 90.59 | 93.55 | 0.9207 | 0.8913 | 0.8396 | 0.9239 |
PU | 80.32 | 78.87 | 81.29 | 0.8008 | 0.7984 | 0.5994 | 0.7697 |
KKI | 88.19 | 87.45 | 88.46 | 0.8796 | 0.8817 | 0.7445 | 0.8256 |
NI | 87.38 | 88.40 | 86.26 | 0.8733 | 0.8296 | 0.7488 | 0.8786 |
Average | 86.96 | 86.33 | 87.39 | 0.8686 | 0.8503 | 0.7331 | 0.8495 |
With ensemble learning | |||||||
NYU | 96.20 | 96.88 | 95.38 | 0.9616 | 0.9400 | 0.9250 | 0.9660 |
PU | 88.79 | 96.67 | 83.50 | 0.8982 | 0.9074 | 0.7801 | 0.8719 |
KKI | 95.05 | 98.90 | 93.66 | 0.9656 | 0.9815 | 0.9033 | 0.9276 |
NI | 93.23 | 98.32 | 87.70 | 0.9302 | 0.9174 | 0.8667 | 0.9374 |
Average | 93.32 | 97.69 | 90.06 | 0.9389 | 0.9366 | 0.8688 | 0.9257 |
Network | NYU (%) | PU (%) | KKI (%) | NI (%) | Average (%) |
Without ensemble learning | |||||
Default-classifier network | 69.00 | 53.69 | 42.82 | 67.67 | 58.29 |
Default-classifier network | 89.83 | 80.26 | 86.84 | 86.79 | 85.93 |
ALFF-based AE network | 91.94 | 80.32 | 88.19 | 87.38 | 86.86 |
With ensemble learning | |||||
Default-classifier network | 75.01 | 52.38 | 35.51 | 75.49 | 59.60 |
Default-classifier network | 95.14 | 85.45 | 91.29 | 92.39 | 91.07 |
ALFF-based AE network | 96.20 | 88.79 | 95.05 | 93.23 | 93.32 |
NYU(%) | PU(%) | KKI(%) | NI(%) | Average(%) | Material | Rawfeatures | Selectedfeatures* | Biomarkerdetection | |
ML | |||||||||
Fusion fMRI (2018) | 52.7 | - | 86.7 | 72.9 | 70.8 | FC | 4005 | - | No |
L1BioSVM (2018) | - | 81.1 | 81.3 | - | 81.2 | FC | 6670 | - | No |
R-Relief (2019) | 70.7 | 68.6 | 81.8 | 76.0 | 74.3 | fALFF | 31 × 37 × 31 | - | No |
DL | |||||||||
3D CNN (2017) | 70.5 | 63.0 | - | 72.8 | 68.8 | MRI image | 90 × 117 × 100 | 1024 | No |
DeepfMRI (2020) | 73.1 | 62.7 | - | 67.9 | 67.9 | BOLD | 90 | 32 | No |
CDAE (2021) | 73.2 | 70.6 | 81.7 | 79.0 | 76.1 | MRI image | 60 × 72× 60 | 5 × 6 × 5 | No |
DVAE (2021) | 62.4 | 67.0 | 78.1 | 68.8 | 69.1 | BOLD | 28,546 | 80 | No |
STAAE (2022) | 93.5 | 92.7 | 90.4 | 91.7 | 92.1 | BOLD | 28,546 | 100 | Yes |
Data augmentation (2023) | 75.6 | 76.5 | 76.0 | - | 76.0 | FC | 13,456 | 1856 | No |
BHT framework | |||||||||
SP-BH (2019) | 96.2 | 95.8 | 86.7 | 91.6 | 92.6 | FC | 4005 | 100 | No |
SP-l2,1-BH (2020) | 99.5 | 96.3 | 100 | 95.8 | 97.9 | FC | 4005 | 50 | No |
AENet (2022) | 99.8 | 99.6 | 99.8 | 99.3 | 99.6 | FC | 4005 | 50 | Yes |
Our | 96.2 | 88.8 | 95.1 | 93.3 | 93.3 | ALFF | 50 | 25 | Yes |
* The number of feature selected in the ML method is not fixed. And the DL method selects the learned high-level features, while the BHT method selects typical features from the raw features. |
Brian region | Ranking order | PU (ADHD-RS) | KKI (CPRS-LV) | NYU (CPRS-LV) | ||||
Name | Abbrev | Corr | P-value | Corr | P-value | Corr | P-value | |
Middle cingulate gyrus (R) | DCG.R | 1 | 0.235 | 0.007 | 0.358 | 0.003 | -0.035 | 0.660 |
Cerebellum Ⅸ (R) | CRBL9.R | 2 | 0.161 | 0.065 | 0.155 | 0.213 | 0.038 | 0.630 |
Amygdala (R) | AMYG.R | 3 | 0.198 | 0.023 | 0.057 | 0.648 | -0.005 | 0.953 |
Thalamus (L) | THA.L | 4 | 0.245 | 0.005 | 0.150 | 0.229 | 0.044 | 0.572 |
Cerebellum Ⅹ (R) | CRBL10.R | 5 | 0.244 | 0.005 | 0.241 | 0.051 | 0.021 | 0.787 |
Cerebellum Crus Ⅱ (L) | CRBL Crus2.L | 6 | 0.245 | 0.005 | 0.259 | 0.036 | 0.128 | 0.102 |
Cerebellum Crus Ⅱ (R) | CRBL Crus2.R | 7 | 0.214 | 0.014 | 0.236 | 0.057 | 0.123 | 0.115 |
Thalamus (R) | THA.R | 8 | 0.270 | 0.002 | 0.184 | 0.139 | 0.043 | 0.580 |
Hippocampus (L) | HIP.L | 9 | 0.274 | 0.001 | 0.273 | 0.027 | 0.021 | 0.786 |
Caudate (R) | CAU.L | 10 | 0.235 | 0.007 | 0.215 | 0.083 | 0.034 | 0.667 |
Site | Age | Female/Male | Control | ADHD | Total |
NYU | 7–18 | 76/140 | 98 | 118 | 216 |
PU | 8–17 | 52/142 | 116 | 78 | 194 |
KKI | 8–13 | 37/46 | 61 | 22 | 83 |
NI | 11–22 | 17/31 | 23 | 25 | 48 |
Region name | Region index | Region name | Region index |
Olfactory cortex | 21–22 | Amygdala | 41–42 |
Insula | 29–30 | Caudate | 71–72 |
Anterior cingulate | 31–32 | Putamen | 73–74 |
Middle cingulate | 33–34 | Pallidum | 75–76 |
Posterior cingulate | 35–36 | Thalamus | 77–78 |
Hippocampus | 37–38 | Cerebellum | 91–108 |
Parahippocampal | 39–40 | Vermis | 109–116 |
Layer size* | |
Dense 1 | (25, 10) |
Dense 2 | (10, 25) |
Dense 3 | (10, 8) |
Dense 4 | (8, 8) |
Dense 5 | (8, 2) |
* The parameters (a, b) describe a dense unit with input size of a and output size of b. |
Site | Accuracy (%) | Sensitivity (%) | Specificity (%) | AUROC | AUPRC | MCC | F1 |
Without ensemble learning | |||||||
NYU | 91.94 | 90.59 | 93.55 | 0.9207 | 0.8913 | 0.8396 | 0.9239 |
PU | 80.32 | 78.87 | 81.29 | 0.8008 | 0.7984 | 0.5994 | 0.7697 |
KKI | 88.19 | 87.45 | 88.46 | 0.8796 | 0.8817 | 0.7445 | 0.8256 |
NI | 87.38 | 88.40 | 86.26 | 0.8733 | 0.8296 | 0.7488 | 0.8786 |
Average | 86.96 | 86.33 | 87.39 | 0.8686 | 0.8503 | 0.7331 | 0.8495 |
With ensemble learning | |||||||
NYU | 96.20 | 96.88 | 95.38 | 0.9616 | 0.9400 | 0.9250 | 0.9660 |
PU | 88.79 | 96.67 | 83.50 | 0.8982 | 0.9074 | 0.7801 | 0.8719 |
KKI | 95.05 | 98.90 | 93.66 | 0.9656 | 0.9815 | 0.9033 | 0.9276 |
NI | 93.23 | 98.32 | 87.70 | 0.9302 | 0.9174 | 0.8667 | 0.9374 |
Average | 93.32 | 97.69 | 90.06 | 0.9389 | 0.9366 | 0.8688 | 0.9257 |
Network | NYU (%) | PU (%) | KKI (%) | NI (%) | Average (%) |
Without ensemble learning | |||||
Default-classifier network | 69.00 | 53.69 | 42.82 | 67.67 | 58.29 |
Default-classifier network | 89.83 | 80.26 | 86.84 | 86.79 | 85.93 |
ALFF-based AE network | 91.94 | 80.32 | 88.19 | 87.38 | 86.86 |
With ensemble learning | |||||
Default-classifier network | 75.01 | 52.38 | 35.51 | 75.49 | 59.60 |
Default-classifier network | 95.14 | 85.45 | 91.29 | 92.39 | 91.07 |
ALFF-based AE network | 96.20 | 88.79 | 95.05 | 93.23 | 93.32 |
NYU(%) | PU(%) | KKI(%) | NI(%) | Average(%) | Material | Rawfeatures | Selectedfeatures* | Biomarkerdetection | |
ML | |||||||||
Fusion fMRI (2018) | 52.7 | - | 86.7 | 72.9 | 70.8 | FC | 4005 | - | No |
L1BioSVM (2018) | - | 81.1 | 81.3 | - | 81.2 | FC | 6670 | - | No |
R-Relief (2019) | 70.7 | 68.6 | 81.8 | 76.0 | 74.3 | fALFF | 31 × 37 × 31 | - | No |
DL | |||||||||
3D CNN (2017) | 70.5 | 63.0 | - | 72.8 | 68.8 | MRI image | 90 × 117 × 100 | 1024 | No |
DeepfMRI (2020) | 73.1 | 62.7 | - | 67.9 | 67.9 | BOLD | 90 | 32 | No |
CDAE (2021) | 73.2 | 70.6 | 81.7 | 79.0 | 76.1 | MRI image | 60 × 72× 60 | 5 × 6 × 5 | No |
DVAE (2021) | 62.4 | 67.0 | 78.1 | 68.8 | 69.1 | BOLD | 28,546 | 80 | No |
STAAE (2022) | 93.5 | 92.7 | 90.4 | 91.7 | 92.1 | BOLD | 28,546 | 100 | Yes |
Data augmentation (2023) | 75.6 | 76.5 | 76.0 | - | 76.0 | FC | 13,456 | 1856 | No |
BHT framework | |||||||||
SP-BH (2019) | 96.2 | 95.8 | 86.7 | 91.6 | 92.6 | FC | 4005 | 100 | No |
SP-l2,1-BH (2020) | 99.5 | 96.3 | 100 | 95.8 | 97.9 | FC | 4005 | 50 | No |
AENet (2022) | 99.8 | 99.6 | 99.8 | 99.3 | 99.6 | FC | 4005 | 50 | Yes |
Our | 96.2 | 88.8 | 95.1 | 93.3 | 93.3 | ALFF | 50 | 25 | Yes |
* The number of feature selected in the ML method is not fixed. And the DL method selects the learned high-level features, while the BHT method selects typical features from the raw features. |
Brian region | Ranking order | PU (ADHD-RS) | KKI (CPRS-LV) | NYU (CPRS-LV) | ||||
Name | Abbrev | Corr | P-value | Corr | P-value | Corr | P-value | |
Middle cingulate gyrus (R) | DCG.R | 1 | 0.235 | 0.007 | 0.358 | 0.003 | -0.035 | 0.660 |
Cerebellum Ⅸ (R) | CRBL9.R | 2 | 0.161 | 0.065 | 0.155 | 0.213 | 0.038 | 0.630 |
Amygdala (R) | AMYG.R | 3 | 0.198 | 0.023 | 0.057 | 0.648 | -0.005 | 0.953 |
Thalamus (L) | THA.L | 4 | 0.245 | 0.005 | 0.150 | 0.229 | 0.044 | 0.572 |
Cerebellum Ⅹ (R) | CRBL10.R | 5 | 0.244 | 0.005 | 0.241 | 0.051 | 0.021 | 0.787 |
Cerebellum Crus Ⅱ (L) | CRBL Crus2.L | 6 | 0.245 | 0.005 | 0.259 | 0.036 | 0.128 | 0.102 |
Cerebellum Crus Ⅱ (R) | CRBL Crus2.R | 7 | 0.214 | 0.014 | 0.236 | 0.057 | 0.123 | 0.115 |
Thalamus (R) | THA.R | 8 | 0.270 | 0.002 | 0.184 | 0.139 | 0.043 | 0.580 |
Hippocampus (L) | HIP.L | 9 | 0.274 | 0.001 | 0.273 | 0.027 | 0.021 | 0.786 |
Caudate (R) | CAU.L | 10 | 0.235 | 0.007 | 0.215 | 0.083 | 0.034 | 0.667 |