1
|
Wahab A, Siddique HR. An update understanding of stemness and chemoresistance of prostate cancer. Expert Rev Anticancer Ther 2025. [PMID: 39935028 DOI: 10.1080/14737140.2025.2466680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION Globally, prostate cancer (CaP) is a leading cause of death and disability among men and a substantial public health burden. Despite advancements in cancer treatment, chemoresistance remains a significant issue in cancer therapy, accounting for the majority of patient relapses and poor survival. Cancer stem cells (CSCs) are considered the main cause of cancer recurrence, chemoresistance, and poor survival of patients. These CSCs acquire stemness and chemoresistance by certain mechanisms such as enhanced DNA repair processes, increased expression of drug efflux pumps, resistance to apoptosis, and altered cell cycle and tumor microenvironment (TME). AREA COVERED We cover the latest developments in this field and give an overview of future research directions. EXPERT OPINION CSCs show dysregulation of several signaling pathways, mostly related to conferring chemoresistance phenotype, such as high drug efflux, apoptotic resistance, quiescent cell cycle, tumor microenvironment, and DNA repair. There are several research articles published on this topic. However, still, this field warrants further investigations to identify the therapeutic molecule that can either chemosensitize CSCs or kill them effectively. This can only be possible when we know the complete mechanisms to comprehend the fundamental causes of cancer stemness and therapy resistance.
Collapse
Affiliation(s)
- Afiya Wahab
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
2
|
Liu Y, Zhu R, Liu D, Hu Y, Xia Q, Liu X, Wang C, Li Y. The characterization of Herbaspirillum huttiense isolated from a uremic patient: virulence and antimicrobial efficacy in the Galleria mellonella model. Lett Appl Microbiol 2025; 78:ovaf012. [PMID: 39919764 DOI: 10.1093/lambio/ovaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 02/09/2025]
Abstract
Herbaspirillum species are typically isolated from plants and are known for their role in crop nitrogen fixation. Recently, they have been found to colonize humans and cause infections. This study aimed to identify and characterize two Herbaspirillum huttiense strains (CLJ01 and CLJ02) isolated from the blood of a uremic patient, with a focus on evaluating their pathogenicity and antibiotic efficacy. The strains were identified using the VITEK2 system, Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry , and 16S rRNA gene sequencing, confirming their identity as H. huttiense. High-throughput sequencing further revealed the presence of the resistance gene arnA, which confers resistance to polymyxin. For the first time, the pathogenicity of H. huttiense was assessed using a Galleria mellonella infection model. The results demonstrated a concentration-dependent virulence, with CLJ01 exhibiting slightly higher pathogenicity. Additionally, meropenem showed significant antimicrobial efficacy in the G. mellonella infection model, particularly under conditions of high bacterial load, indicating strong therapeutic potential. In conclusion, this study provides experimental evidence supporting the correct diagnosis and treatment of H. huttiense infections. Furthermore, the findings underscore the importance of accurately identifying rare pathogens in clinical settings for effective treatment.
Collapse
Affiliation(s)
- Yuchun Liu
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Rui Zhu
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Dongmei Liu
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Yue Hu
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Qing Xia
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Xinwei Liu
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Chunxia Wang
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| | - Yongwei Li
- The Second Affiliated Hospital of Henan University of Chinese Medicine, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou 450002, China
| |
Collapse
|
3
|
Wu W, Guo Z, Zhang J, Li W, Liu C, Jiang B, Su Y. Integration of transcriptomics and metabolomics reveals the mechanism of enrofloxacin resistance in Aeromonas schubertii. Microb Pathog 2025; 199:107262. [PMID: 39730098 DOI: 10.1016/j.micpath.2024.107262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/18/2024] [Accepted: 12/24/2024] [Indexed: 12/29/2024]
Abstract
Aeromonas schubertii infections has caused severe economic losses in aquaculture in China. In this study, we first induced enrofloxacin (ENR) resistance in A. schubertii strains and then analyzed the mechanisms of drug resistance using transcriptomics and metabolomics. We found that the minimal inhibitory concentration (MIC) was 0.03125 μg/mL for the sensitive strain (WL23S) and 32 μg/mL for the resistant strain (WL23R), which is a 1024-fold increase. After 40 serial passages, the WL23R strain maintained a MIC of 32 μg/mL, even in the absence of ENR-induced stress. Notably, it had also developed resistance to several other antibiotics, such as neomycin sulfate and flumequine. There was no significant difference in the growth rates of the two strains, highlighting the strong adaptability and growth characteristics of the WL23R strain. Comparison of the transcriptome data between the WL23R and WL23S strains identified 579 differentially expressed genes. Expression of the efflux pump-related genes (e.g., acrA, acrB, pstB, pstC, pstS) was significantly upregulated in the WL23R strain (P < 0.05). The highest enrichment of differential genes in the Gene Ontology analysis was in the catabolism of various amino acids, and that in the Kyoto Encyclopedia of Genes and Genomes pathway was in ATP-binding cassette (ABC) transport. Comparison of the metabolomics data between the WL23R and WL23S strains revealed 1, 059 differentially expressed metabolites. Metabolomics analysis revealed the impact of drug resistance on the levels of amino acids, the activity of amino acid biosynthesis/metabolism pathways, and the ABC transport protein pathway, which confirmed the transcriptomics results. The joint analysis results showed that ABC transporters were most prominent in the shared pathways between enriched differentially expressed genes and metabolites. To further validate the resistance mechanism of A. schubertii, we exposed the WL23R strain to the efflux pump inhibitor carbonyl cyanide 3-chlorophenylhydrazone. The minimal inhibitory concentration of the induced resistant strain decreased by 4-fold after the addition of the inhibitor, indicating the overexpression of active efflux pumps in WL23R. Our results indicate that the efflux system and ABC transporters play crucial roles during the development of multidrug resistance in A. schubertii. This study will serve as an important reference for understanding bacterial resistance to quinolones and multidrug resistance in aquatic environments.
Collapse
Affiliation(s)
- Wenhao Wu
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Zihong Guo
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Jiahao Zhang
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Wei Li
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Chun Liu
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China
| | - Biao Jiang
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China.
| | - Youlu Su
- Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, Guangdong, China.
| |
Collapse
|
4
|
Wimalasekara RL, White D, Kumar A. Targeting Acinetobacter baumannii resistance-nodulation-division efflux pump transcriptional regulators to combat antimicrobial resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:4. [PMID: 39863717 PMCID: PMC11762787 DOI: 10.1038/s44259-024-00074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Regulatory elements controlling gene expression fine-tune bacterial responses to environmental cues, including antimicrobials, to optimize survival. Acinetobacter baumannii, a pathogen notorious for antimicrobial resistance, relies on efficient efflux systems. Though the role of efflux systems in antibiotic expulsion are well recognized, the regulatory mechanisms controlling their expression remain understudied. This review explores the current understanding of these regulators, aiming to inspire strategies to combat bacterial resistance and improve therapeutic outcomes.
Collapse
Affiliation(s)
| | - Dawn White
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Ayush Kumar
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
5
|
Qiu L, Guo X, Shim H, Hao T, Liang Z, Wang S, Lu Z, Lu Q, He Z. Unveiling triclosan biodegradation: Novel metabolic pathways, genomic insights, and global environmental adaptability of Pseudomonas sp. strain W03. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137313. [PMID: 39862779 DOI: 10.1016/j.jhazmat.2025.137313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
The polychlorinated aromatic antimicrobial agent triclosan (TCS) is widely used to indiscriminately and rapidly kill microorganisms. The global use of TCS has led to widespread environmental contamination, posing significant threats to ecosystem and human health. Here we reported a newly isolated Pseudomonas sp. W03 for degrading TCS metabolically at concentrations up to 10 mg/L. This strain exhibited optimal degradation activity at 30°C and pH 7.0, and retained substantial activity at pH 4.0, although it was sensitive to alkaline conditions. Genomic analysis of strain W03 revealed a circular chromosome comprising 6075,907 bp with a GC content of 65.08 %. A novel TCS degradation pathway, involving dechlorination, oxidation, ether bond fission, and reoxidation processes, was identified. Also, the study mapped the global distribution of analogous Pseudomonas using 16S rRNA gene sequences, revealing their widespread presence in diverse aquatic environments, with a significant abundance in wastewater systems. These findings indicated that these bacteria play a critical ecological role in both natural and engineered environments, particularly in the degradation of organic pollutants. This study enhances our understanding of microbial degradation of emerging contaminants and presents a promising candidate for bioremediation strategies aimed at mitigating TCS-related water pollution.
Collapse
Affiliation(s)
- Lan Qiu
- Marine Synthetic Ecology Research Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China; Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Xiaoyuan Guo
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Hojae Shim
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Tianwei Hao
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, 999078, Macao
| | - Zhiwei Liang
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanquan Wang
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhenmei Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qihong Lu
- Department of Environmental Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China.
| | - Zhili He
- Marine Synthetic Ecology Research Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Science, Sun Yat-sen University, Zhuhai 519080, China.
| |
Collapse
|
6
|
Ikhane AO, Osunsanmi FO, Mosa RA, Opoku AR. Antibacterial Potential of Crude Extracts from Cylindrospermum alatosporum NR125682 and Loriellopsis cavernicola NR117881. Microorganisms 2025; 13:211. [PMID: 39858979 PMCID: PMC11767720 DOI: 10.3390/microorganisms13010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The challenges of antimicrobial resistance (AMR) to human health have pushed for the discovery of a new antibiotics agent from natural products. Cyanobacteria are oxygen-producing photosynthetic prokaryotes found in a variety of water habitats. Secondary metabolites are produced by cyanobacteria to survive extreme environmental stress factors, including microbial competition. This study presents the antibacterial activity and mechanism of the crude extracts from Cylindrospermum alatosporum NR125682 (A) and Loriellopsis cavernicola NR117881 (B) isolated from freshwater. The cyanobacteria were identified through 16S rRNA sequencing. Crude extracts were sequentially prepared using hexane, dichloromethane, and ethanol consistently. The minimum inhibition concentration (MIC), minimum bactericidal concentration (MBC) using the CSLI microdilution test protocol, and crude extract potential to inhibit the growth of the tested clinical bacteria strains were evaluated. The mechanism of action of the extracts including membrane damage, efflux pump, β-lactamase activity, DNA degradation, and extract-drug interaction was investigated using standard procedures. The hexane extract of B performed the best with a MIC (0.7-1.41 mg/mL) and MBC (1.41-2.81 mg/mL) range. All the crude extracts inhibited efflux pump activity against the bacteria tested. However, the extracts poorly inhibited β-lactamase. The ethanol extract of B exhibited the most appreciable antibacterial activity. The dichloromethane extract of B showed the highest significant DNA degradation potential, when compared with other samples. The extracts exhibited synergism when combined with erythromycin against some test bacteria, indicating primary microbial activity through membrane interactions. Hence, this study demonstrates the significance of cyanobacteria for antibiotic development.
Collapse
Affiliation(s)
- Albert Olufemi Ikhane
- Department of Biochemistry and Microbiology, University of Zululand, Richards Bay 3886, South Africa;
| | | | - Rebamang Anthony Mosa
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield 0002, South Africa;
| | - Andrew Rowland Opoku
- Department of Biochemistry and Microbiology, University of Zululand, Richards Bay 3886, South Africa;
| |
Collapse
|
7
|
Sharma D, Gautam S, Singh S, Srivastava N, Khan AM, Bisht D. Unveiling the nanoworld of antimicrobial resistance: integrating nature and nanotechnology. Front Microbiol 2025; 15:1391345. [PMID: 39850130 PMCID: PMC11754303 DOI: 10.3389/fmicb.2024.1391345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
A significant global health crisis is predicted to emerge due to antimicrobial resistance by 2050, with an estimated 10 million deaths annually. Increasing antibiotic resistance necessitates continuous therapeutic innovation as conventional antibiotic treatments become increasingly ineffective. The naturally occurring antibacterial, antifungal, and antiviral compounds offer a viable alternative to synthetic antibiotics. This review presents bacterial resistance mechanisms, nanocarriers for drug delivery, and plant-based compounds for nanoformulations, particularly nanoantibiotics (nAbts). Green synthesis of nanoparticles has emerged as a revolutionary approach, as it enhances the effectiveness, specificity, and transport of encapsulated antimicrobials. In addition to minimizing systemic side effects, these nanocarriers can maximize therapeutic impact by delivering the antimicrobials directly to the infection site. Furthermore, combining two or more antibiotics within these nanoparticles often exhibits synergistic effects, enhancing the effectiveness against drug-resistant bacteria. Antimicrobial agents are routinely obtained from secondary metabolites of plants, including essential oils, phenols, polyphenols, alkaloids, and others. Integrating plant-based antibacterial agents and conventional antibiotics, assisted by suitable nanocarriers for codelivery, is a potential solution for addressing bacterial resistance. In addition to increasing their effectiveness and boosting the immune system, this synergistic approach provides a safer and more effective method of tackling future bacterial infections.
Collapse
Affiliation(s)
- Devesh Sharma
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, India
- School of Studies in Biochemistry, Jiwaji University, Gwalior, India
| | - Sakshi Gautam
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, India
| | - Sakshi Singh
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, India
| | - Nalini Srivastava
- School of Studies in Biochemistry, Jiwaji University, Gwalior, India
| | - Abdul Mabood Khan
- Division of Clinical Trials and Implementation Research, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, India
| | - Deepa Bisht
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, India
| |
Collapse
|
8
|
Azeem K, Fatima S, Ali A, Ubaid A, Husain FM, Abid M. Biochemistry of Bacterial Biofilm: Insights into Antibiotic Resistance Mechanisms and Therapeutic Intervention. Life (Basel) 2025; 15:49. [PMID: 39859989 PMCID: PMC11767195 DOI: 10.3390/life15010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Biofilms, composed of structured communities of bacteria embedded in a self-produced extracellular matrix, pose a significant challenge due to their heightened resistance to antibiotics and immune responses. This review highlights the mechanisms underpinning antibiotic resistance within bacterial biofilms, elucidating the adaptive strategies employed by microorganisms to withstand conventional antimicrobial agents. This encompasses the role of the extracellular matrix, altered gene expression, and the formation of persister cells, contributing to the recalcitrance of biofilms to eradication. A comprehensive understanding of these resistance mechanisms provides a for exploring innovative therapeutic interventions. This study explores promising avenues for future research, emphasizing the necessity of uncovering the specific genetic and phenotypic adaptations occurring within biofilms. The identification of vulnerabilities in biofilm architecture and the elucidation of key biofilm-specific targets emerge as crucial focal points for the development of targeted therapeutic strategies. In addressing the limitations of traditional antibiotics, this review discusses innovative therapeutic approaches. Nanomaterials with inherent antimicrobial properties, quorum-sensing inhibitors disrupting bacterial communication, and bacteriophages as biofilm-specific viral agents are highlighted as potential alternatives. The exploration of combination therapies, involving antimicrobial agents, biofilm-disrupting enzymes, and immunomodulators, is emphasized to enhance the efficacy of existing treatments and overcome biofilm resilience.
Collapse
Affiliation(s)
- Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Sadaf Fatima
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
- Clinical Biochemistry Laboratory, Department of Biochemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Ubaid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| |
Collapse
|
9
|
Zhang S, Shu Y, Yang Z, Zhong Z, Wang M, Jia R, Chen S, Liu M, Zhu D, Zhao X, Wu Y, Yang Q, Huang J, Ou X, Sun D, Tian B, Wu Z, He Y, Cheng A. Decoding the enigma: unveiling the transmission characteristics of waterfowl-associated bla NDM-5-positive Escherichia coli in select regions of China. Front Microbiol 2024; 15:1501594. [PMID: 39717269 PMCID: PMC11663885 DOI: 10.3389/fmicb.2024.1501594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Escherichia coli (E. coli) serves as a critical indicator microorganism for assessing the prevalence and dissemination of antibiotic resistance, notably harboring various antibiotic-resistant genes (ARGs). Among these, the emergence of the bla NDM gene represents a significant threat to public health, especially since carbapenem antibiotics are vital for treating severe infections caused by Gram-negative bacteria. This study aimed to characterize the antibiotic resistance features of bla NDM-5-positive E. coli strains isolated from waterfowl in several regions of China and elucidate the dissemination patterns of the bla NDM-5 gene. We successfully isolated 103 bla NDM-5-positive E. coli strains from 431 intestinal fecal samples obtained from waterfowl across five provincial-level units in China, with all strains exhibiting multidrug resistance (MDR). Notably, the bla NDM-5 gene was identified on plasmids, which facilitate efficient and stable horizontal gene transfer (HGT). Our adaptability assays indicated that while the bla NDM-5-positive plasmid imposed a fitness cost on the host bacteria, the NDM-5 protein was successfully induced and purified, exhibiting significant enzymatic activity. One strain, designated DY51, exhibited a minimum inhibitory concentration (MIC) for imipenem of 4 mg/L, which escalated to 512 mg/L following exposure to increasing imipenem doses. This altered strain demonstrated stable resistance to imipenem alongside improved adaptability, correlating with elevated relative expression levels of the bla NDM-5 and overexpression of efflux pumps. Collectively, this study highlights the horizontal dissemination of the bla NDM-5 plasmid among E. coli strains, confirms the associated fitness costs, and provides insights into the mechanisms underlying the stable increase in antibiotic resistance to imipenem. These findings offer a theoretical framework for understanding the dissemination dynamics of bla NDM-5 in E. coli, which is essential for developing effective strategies to combat carbapenem antibiotic resistance.
Collapse
Affiliation(s)
- Shaqiu Zhang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Yanxi Shu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhechen Yang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Renyong Jia
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Shun Chen
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Mafeng Liu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Xinxin Zhao
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Ying Wu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Qiao Yang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Juan Huang
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Xumin Ou
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Di Sun
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Bin Tian
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Zhen Wu
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Yu He
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| | - Anchun Cheng
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the P.R. China, Chengdu, China
| |
Collapse
|
10
|
He L, Kim Y, Kim S, Lee MH, Yu JM. Unveiling Antibiotic Resistance: Genome Sequencing of Streptomycin-Resistant Erwinia amylovora Isolate. Microorganisms 2024; 12:2494. [PMID: 39770697 PMCID: PMC11728212 DOI: 10.3390/microorganisms12122494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Erwinia amylovora, the causal agent of fire blight, poses a serious threat to several rosaceous plants, especially apples and pears. In this study, a spontaneous streptomycin-resistant E. amylovora strain (EaSmR) was isolated under laboratory conditions. Compared with the parental strain TS3128, the EaSmR strain exhibited high resistance to streptomycin (>100,000 µg/mL) and showed a significant reduction in both swimming and swarming motility. To investigate the mechanisms underlying streptomycin resistance, the genome of EaSmR was sequenced, and four single nucleotide variants (SNVs) were identified in comparison with the EaSmR genome with TS3128. Two genes in EaSmR were found to contain SNVs relative to TS3128, including a point mutation at codon 43 in the rpsL gene, the primary target of streptomycin, which was identified as the cause of the resistance. Additionally, three other point mutations were detected within the gene encoding type I methionyl aminopeptidase (MetAP1), resulting in an amino acid substitution from serine to valine (S76V). Furthermore, we analyzed the nonsynonymous single nucleotide variants (nsSNVs) between the EaSmR isolate and the reference type strain, CFBP1430. A total of 111 nsSNVs were found in EaSmR, including three stop-gain mutations, across 102 genes, which likely account for potential differences between the Korean strain TS3128 (EaSmR) and the reference strain CFBP1430. Whole-genome sequencing of EaSmR reveals significant genetic changes and provides valuable insights into the role of single nucleotide variants in antibiotic resistance and altered physiological traits. As the first report of a laboratory-induced, streptomycin-resistant E. amylovora strain from South Korea, this study provides essential insights into resistance mechanisms and highlights key genomic differences that may contribute to the unique characteristics of the Korean strain, establishing a valuable foundation for future disease management strategies.
Collapse
Affiliation(s)
- Lin He
- Department of Applied Biology, Chungnam National University, Daejeon 34134, Republic of Korea; (L.H.); (Y.K.); (S.K.)
| | - Yuna Kim
- Department of Applied Biology, Chungnam National University, Daejeon 34134, Republic of Korea; (L.H.); (Y.K.); (S.K.)
| | - Seohyun Kim
- Department of Applied Biology, Chungnam National University, Daejeon 34134, Republic of Korea; (L.H.); (Y.K.); (S.K.)
| | - Mi-Hyun Lee
- Crop Protection Division, National Institute of Agricultural Sciences, Wanju 55365, Republic of Korea;
| | - Jun Myoung Yu
- Department of Applied Biology, Chungnam National University, Daejeon 34134, Republic of Korea; (L.H.); (Y.K.); (S.K.)
| |
Collapse
|
11
|
Di Maso AM, Ruiz C. Physiological Effects of TolC-Dependent Multidrug Efflux Pumps in Escherichia coli: Impact on Motility and Growth Under Stress Conditions. Microbiologyopen 2024; 13:e70006. [PMID: 39529380 PMCID: PMC11554990 DOI: 10.1002/mbo3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Enterobacteriaceae possess eight TolC-dependent multidrug efflux pumps: AcrAB-TolC, AcrAD-TolC, AcrEF-TolC, MdtEF-TolC, MdtABC-TolC, EmrAB-TolC, EmrYK-TolC, and MacAB-TolC, which efflux bile salts, antibiotics, metabolites, or other compounds. However, our understanding of their physiological roles remains limited, especially for less-studied pumps like EmrYK-TolC. In this study, we tested the effects on swimming motility and growth under stress conditions of Escherichia coli mutants individually deleted for each inner-membrane transporter component of all eight TolC-dependent pumps, a mutant deleted for the AcrB-accessory protein AcrZ, and a mutant simultaneously deleted for all eight pumps (ΔtolC). We found that all mutants tested, except the ΔemrY and ΔacrZ mutants, displayed increased swimming motility. Additionally, the loss of each individual TolC-dependent pump or AcrZ did not reduce growth and sometimes even enhanced it compared to the parental strain under various growth conditions: temperature (LB at 25, 30, 37, and 42°C), pH (LB at pH 6.0, 7.4, and 9.0; and LB buffered to pH 6.0, 7.4, and 8.25), LB with limited air exchange, and nutritional stress (M9-glucose or M9-glycerol). In contrast, the ΔtolC mutant grew significantly slower than the parental strain under all conditions tested except in LB-TRIS pH 7.4 and LB with limited air exchange. Overall, these findings indicate that while individual TolC-dependent pumps are generally dispensable for growth under many stress conditions in the absence of antimicrobials, possibly due to their partially overlapping substrate profiles, TolC-dependent efflux is required for maximal growth under most conditions.
Collapse
Affiliation(s)
- Amanda M. Di Maso
- Department of BiologyCalifornia State University NorthridgeNorthridgeCaliforniaUSA
| | - Cristian Ruiz
- Department of BiologyCalifornia State University NorthridgeNorthridgeCaliforniaUSA
| |
Collapse
|
12
|
Salmani-Zarchi H, Mousavi-Sagharchi SMA, Sepahdoost N, Ranjbar-Jamalabadi M, Gross JD, Jooya H, Samadi A. Antimicrobial Feature of Nanoparticles in the Antibiotic Resistance Era: From Mechanism to Application. Adv Biomed Res 2024; 13:113. [PMID: 39717242 PMCID: PMC11665187 DOI: 10.4103/abr.abr_92_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 12/25/2024] Open
Abstract
The growth of nanoscale sciences enables us to define and design new methods and materials for a better life. Health and disease prevention are the main issues in the human lifespan. Some nanoparticles (NPs) have antimicrobial properties that make them useful in many applications. In recent years, NPs have been used as antibiotics to overcome drug resistance or as drug carriers with antimicrobial features. They can also serve as antimicrobial coatings for implants in different body areas. The antimicrobial feature of NPs is based on different mechanisms. For example, the oxidative functions of NPs can inhibit nucleic acid replication and destroy the microbial cell membrane as well as interfere with their cellular functions and biochemical cycles. On the other hand, NPs can disrupt the pathogens' lifecycle by interrupting vital points of their life, such as virus uncoating and entry into human cells. Many types of NPs have been tested by different scientists for these purposes. Silver, gold, copper, and titanium have shown the most ability to inhibit and remove pathogens inside and outside the body. In this review, the authors endeavor to comprehensively describe the antimicrobial features of NPs and their applications for different biomedical goals.
Collapse
Affiliation(s)
- Hamed Salmani-Zarchi
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Nafise Sepahdoost
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdieh Ranjbar-Jamalabadi
- Department of Polymer Engineering, Faculty of Chemical Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Jeffrey D. Gross
- ReCELLebrate Regenerative Medicine Clinic, Henderson, Nevada, USA
| | - Hossein Jooya
- Biochemistry Group, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ali Samadi
- Department of Basic Sciences, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
13
|
Silva TO, Bulla ACS, Teixeira BA, Gomes VMS, Raposo T, Barbosa LS, da Silva ML, Moreira LO, Olsen PC. Bacterial efflux pump OMPs as vaccine candidates against multidrug-resistant Gram-negative bacteria. J Leukoc Biol 2024; 116:1237-1253. [PMID: 39011942 DOI: 10.1093/jleuko/qiae154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
The emergence and propagation of bacteria resistant to antimicrobial drugs is a serious public health threat worldwide. The current antibacterial arsenal is becoming obsolete, and the pace of drug development is decreasing, highlighting the importance of investment in alternative approaches to treat or prevent infections caused by antimicrobial-resistant bacteria. A significant mechanism of antimicrobial resistance employed by Gram-negative bacteria is the overexpression of efflux pumps that can extrude several compounds from the bacteria, including antimicrobials. The overexpression of efflux pump proteins has been detected in several multidrug-resistant Gram-negative bacteria, drawing attention to these proteins as potential targets against these pathogens. This review will focus on the role of outer membrane proteins from efflux pumps as potential vaccine candidates against clinically relevant multidrug-resistant Gram-negative bacteria, discussing advantages and pitfalls. Additionally, we will explore the relevance of efflux pump outer membrane protein diversity and the possible impact of vaccination on microbiota.
Collapse
Affiliation(s)
- Thaynara O Silva
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 05, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 07, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Ana Carolina S Bulla
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Bárbara A Teixeira
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 05, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Vinnicius Machado Schelk Gomes
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal do Rio de Janeiro, Avenida São José do Barreto, 764. Centro, Macaé, RJ, 27965-045, Brazil
| | - Thiago Raposo
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 05, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luiza S Barbosa
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 05, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 07, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Manuela Leal da Silva
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, 21040-900, Brazil
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal do Rio de Janeiro, Avenida São José do Barreto, 764. Centro, Macaé, RJ, 27965-045, Brazil
| | - Lilian O Moreira
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 07, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Priscilla C Olsen
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, Bloco A 2º Andar sala 05, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| |
Collapse
|
14
|
Zhao Y, Xu H, Wang H, Wang P, Chen S. Multidrug resistance in Pseudomonas aeruginosa: genetic control mechanisms and therapeutic advances. MOLECULAR BIOMEDICINE 2024; 5:62. [PMID: 39592545 PMCID: PMC11599538 DOI: 10.1186/s43556-024-00221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Pseudomonas aeruginosa is a significant opportunistic pathogen, and its complex mechanisms of antibiotic resistance pose a challenge to modern medicine. This literature review explores the advancements made from 1979 to 2024 in understanding the regulatory networks of antibiotic resistance genes in Pseudomonas aeruginosa, with a particular focus on the molecular underpinnings of these resistance mechanisms. The review highlights four main pathways involved in drug resistance: reducing outer membrane permeability, enhancing active efflux systems, producing antibiotic-inactivating enzymes, and forming biofilms. These pathways are intricately regulated by a combination of genetic regulation, transcriptional regulators, two-component signal transduction, DNA methylation, and small RNA molecules. Through an in-depth analysis and synthesis of existing literature, we identify key regulatory elements mexT, ampR, and argR as potential targets for novel antimicrobial strategies. A profound understanding of the core control nodes of drug resistance offers a new perspective for therapeutic intervention, suggesting that modulating these elements could potentially reverse resistance and restore bacterial susceptibility to antibiotics. The review looks forward to future research directions, proposing the use of gene editing and systems biology to further understand resistance mechanisms and to develop effective antimicrobial strategies against Pseudomonas aeruginosa. This review is expected to provide innovative solutions to the problem of drug resistance in infectious diseases.
Collapse
Affiliation(s)
- Yuanjing Zhao
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Haoran Xu
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Hui Wang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ping Wang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| | - Simin Chen
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
15
|
Silva TO, Teixeira BA, Costa LVS, Barbosa LS, do Nascimento LC, Fanticelli JGC, Rotilho C, Branco RVC, Silva LS, Ferreira ME, Costa TL, Monteiro SV, Dos Santos Abreu J, Rajsfus BF, Bulla ACS, Carneiro J, Allonso D, Salgado DR, Echevarria-Lima J, da Silva ML, Moreira LO, Olsen PC. The Escherichia coli TolC efflux pump protein is immunogenic and elicits protective antibodies. J Leukoc Biol 2024; 116:1398-1411. [PMID: 39278634 DOI: 10.1093/jleuko/qiae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
Antimicrobial resistance is an increasing worldwide public health burden that threatens to make existent antimicrobials obsolete. An important mechanism of antimicrobial resistance is the overexpression of efflux pumps, which reduce the intracellular concentration of antimicrobials. TolC is the outer membrane protein of an efflux pump that has gained attention as a therapeutic target. Little is known about the immune response against TolC. Here, we evaluated the immune response against TolC from Escherichia coli. TolC in silico epitope prediction showed several residues that could bind to human antibodies, and we showed that human plasma presented higher titers of anti-TolC IgG and IgA, than IgM. E. coli recombinant TolC protein stimulated macrophages in vitro to produce nitric oxide, as well as interleukin-6 and tumor necrosis factor α, assessed by Griess assay and enzyme-linked immunosorbent assay, respectively. Immunization of mice with TolC intraperitoneally and an in vitro restimulation led to increased T cell proliferation and interferon γ production, evaluated by flow cytometry and enzyme-linked immunosorbent assay, respectively. TolC mouse immunization stimulated anti-TolC IgM and IgG production, with higher levels of IgG1 and IgG2, among the IgG subclasses. Anti-TolC murine antibodies could bind to live E. coli and increase bacterial uptake and elimination by macrophages in vitro. Intraperitoneal or intranasal, but not oral, immunizations with inactivated E. coli also led to anti-TolC antibody production. Finally, TolC immunization increased mouse survival rates to antimicrobial-sensitive or resistant E. coli infection. Our results showed that TolC is immunogenic, leading to the production of protective antibodies against E. coli, reinforcing its value as a therapeutic target.
Collapse
Affiliation(s)
- Thaynara O Silva
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar sala - 07, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Bárbara A Teixeira
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Leon V S Costa
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Luiza S Barbosa
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar sala - 07, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Lucas C do Nascimento
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - João G C Fanticelli
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Caroline Rotilho
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Rafael V C Branco
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Lucas S Silva
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Maria E Ferreira
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Thais L Costa
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Sanderson V Monteiro
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Juliana Dos Santos Abreu
- Laboratório de Imunologia Básica e Aplicada, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS, Bloco I, 2° andar - sala 43, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Bia F Rajsfus
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Ana Carolina S Bulla
- Programa de Pós-graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, Manguinhos, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Jordanna Carneiro
- Centro de Terapia Intensiva, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 255 - Cidade Universitária, Ilha do Fundão, 21941-617 Rio de Janeiro, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Diamantino R Salgado
- Centro de Terapia Intensiva, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 255 - Cidade Universitária, Ilha do Fundão, 21941-617 Rio de Janeiro, Brazil
| | - Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, CCS, Bloco I, 2° andar - sala 43, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Manuela Leal da Silva
- Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal do Rio de Janeiro, Avenida Amaro Reinaldo dos Santos Silva, 764, São José do Barreto, CEP 27965-045 Macaé, Brazil
| | - Lilian O Moreira
- Laboratório de Bacteriologia e Imunologia Clínica, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar sala - 07, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratório de Estudos em Imunologia, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Bloco A, 2° Andar - sala 05, Cidade Universitária, Ilha do Fundão, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Anedda E, Alexa EA, Farrell ML, Croffie M, Madigan G, Morris D, Burgess CM. Comparison of antimicrobial resistant Enterobacterales isolates from the dairy production environment in low and high zinc containing regions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:175905. [PMID: 39218095 DOI: 10.1016/j.scitotenv.2024.175905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Heavy metals occur naturally in the environment, and their concentration varies in soil across different regions. However, the presence of heavy metals may influence the antimicrobial resistance (AMR) in bacterial populations. Therefore, the objective of this study was to investigate and characterise the antimicrobial resistance profiles of Enterobacterales in soil and bovine milk filters from high and low zinc-containing regions in Ireland. In total, 50 soil samples and 29 milk filters were collected from two geographic locations with varying soil zinc concentrations. Samples were cultured for the enumeration and detection of Enterobacterales. Specifically, extended-spectrum beta-lactamase-producing Enterobacterales, carbapenem-resistant Enterobacterales and ciprofloxacin-resistant Enterobacterales were isolated using selective media. Species identification was performed using MALDI-TOF. The phenotypic resistance profiles of selected Enterobacterales were determined by disk diffusion testing, following EUCAST and CLSI criteria; while, the genotypic resistance profiles of the same isolates were determined by whole genome sequencing (WGS). Heavy metal concentrations were also measured for all soil samples. A total of 40 antimicrobial resistant Enterobacterales were identified in soil (n = 31) and milk filters (n = 9). The predominant species detected in the high zinc-containing region was Escherichia coli in both sample types (soil n = 10, milk filters n = 2), while in the low zinc-containing region Serratia fonticola was predominant in soil samples (n = 8) and E. coli in milk filters (n = 4). Ten E. coli isolates identified from soil samples in the high zinc-containing region were multidrug resistant, showing resistance to all the antimicrobials tested, except for carbapenems. The WGS findings confirmed the phenotypic resistance results. Moreover, zinc resistance-associated genes and genes encoding for efflux pumps were identified. The current study revealed distinct phenotypic resistance profiles of Enterobacterales in low and high zinc-containing regions, and highlighted the benefit of utilising milk filters for AMR surveillance in dairy production.
Collapse
Affiliation(s)
- E Anedda
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland; Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland
| | - E A Alexa
- School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| | - M L Farrell
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland; School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - M Croffie
- Environment, Soils and Land Use Department, Teagasc Johnstown Castle Research Centre, Wexford, Ireland
| | - G Madigan
- Bacteriology/Parasitology Division, Department of Agriculture, Food and the Marine, Backweston Complex, Celbridge, Ireland
| | - D Morris
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland; Centre for One Health, Ryan Institute, University of Galway, Ireland.
| | - C M Burgess
- Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland.
| |
Collapse
|
17
|
Anedda E, Ekhlas D, Alexa E, Farrell ML, Gaffney MT, Madigan G, Morris D, Burgess CM. Characterization of antimicrobial resistant Enterobacterales isolated from spinach and soil following zinc amendment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124774. [PMID: 39178936 DOI: 10.1016/j.envpol.2024.124774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
Antimicrobial resistant bacteria can occur in the primary food production environment. The emergence and dissemination of antimicrobial resistance (AMR) in the environment can be influenced by several factors, including the presence of heavy metals. The aim of this study was to examine the presence and characteristics of antimicrobial resistant Enterobacterales in soils and spinach grown in soils with and without zinc amendment. A total of 160 samples (92 soil and 68 spinach) were collected from two locations, in which some plots had been amended with zinc. Samples were cultured on selective agars for detection of extended-spectrum beta-lactamase-producing Enterobacterales (ESBL), carbapenem-resistant Enterobacterales and ciprofloxacin-resistant Enterobacterales. Samples were also cultured for enumeration of total Enterobacterales. Isolates were identified by MALDI-TOF. Antimicrobial susceptibility testing was carried out in accordance with EUCAST and CLSI criteria. The whole genome sequence (WGS) of selected isolates was determined. Inductively coupled plasma atomic emission spectrometry was also performed on soil samples in order to measure the concentration of zinc. In total 20 antimicrobial resistant Enterobacterales were isolated from the soil (n = 8) and spinach samples (n = 12). In both sample types, Serratia fonticola (n = 16) was the dominant species, followed by Escherichia coli (n = 1), Citrobacter freundii (n = 1) and Morganella morganii (n = 1) detected in spinach samples, and Enterobacter cloacae (n = 1) detected in a soil sample. The WGS identified genes conferring resistance to different antimicrobials in agreement with the phenotypic results; 14 S. fonticola isolates were confirmed as ESBL producers and harboured the blaFONA gene. Genes that encoded for zinc resistance and multidrug efflux pumps, transporters that can target both antimicrobials and heavy metals, were also identified. Overall, the findings of this study suggest the presence of zinc did not influence the AMR Enterobacterales in soil or spinach samples.
Collapse
Affiliation(s)
- E Anedda
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland; Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland
| | - D Ekhlas
- Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - E Alexa
- Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland
| | - M L Farrell
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland
| | - M T Gaffney
- Horticultural Development Department, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - G Madigan
- Bacteriology/Parasitology Division, Department of Agriculture, Food and the Marine, Backweston Complex, Celbridge, Ireland
| | - D Morris
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Ireland; Centre for One Health, Ryan Institute, University of Galway, Ireland
| | - C M Burgess
- Food Safety Department, Teagasc Food Research Centre Ashtown, Dublin, Ireland.
| |
Collapse
|
18
|
Krzyżek P. Helicobacter pylori Efflux Pumps: A Double-Edged Sword in Antibiotic Resistance and Biofilm Formation. Int J Mol Sci 2024; 25:12222. [PMID: 39596287 PMCID: PMC11594842 DOI: 10.3390/ijms252212222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Helicobacter pylori is a major pathogen associated with various gastric diseases. Despite decades of research, the treatment of H. pylori remains challenging. One of the primary mechanisms contributing to failures of therapies targeting this bacterium is genetic mutations in drug target sites, although the growing body of scientific data highlights that efflux pumps may also take part in this process. Efflux pumps are proteinaceous transporters actively expelling antimicrobial agents from the interior of the targeted cells and reducing the intracellular concentration of these compounds. Considering that efflux pumps contribute to both antimicrobial resistance and biofilm formation, an in-depth understanding of their properties may constitute a cornerstone in the development of novel therapeutics against H. pylori. In line with this, the aim of the current review is to describe the multitude of efflux pumps produced by H. pylori and present the data describing the involvement of these proteins in tolerance and/or resistance to various classes of antimicrobial substances.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
19
|
Caioni G, Reyes CP, Laurenti D, Chiaradia C, Dainese E, Mattioli R, Di Risola D, Santavicca E, Francioso A. Biochemistry and Future Perspectives of Antibiotic Resistance: An Eye on Active Natural Products. Antibiotics (Basel) 2024; 13:1071. [PMID: 39596764 PMCID: PMC11591525 DOI: 10.3390/antibiotics13111071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Antibiotic resistance poses a serious threat to the current healthcare system, negatively impacting the effectiveness of many antimicrobial treatments. The situation is exacerbated by the widespread overuse and abuse of available antibiotics, accelerating the evolution of resistance. Thus, there is an urgent need for novel approaches to therapy to overcome established resistance mechanisms. Plants produce molecules capable of inhibiting bacterial growth in various ways, offering promising paths for the development of alternative antibiotic medicine. This review emphasizes the necessity of research efforts on plant-derived chemicals in the hopes of finding and creating novel drugs that can successfully target resistant bacterial populations. Investigating these natural chemicals allows us to improve our knowledge of novel antimicrobial pathways and also expands our antibacterial repertoire with novel molecules. Simultaneously, it is still necessary to utilize present antibiotics sparingly; prudent prescribing practices must be encouraged to extend the effectiveness of current medications. The combination of innovative drug research and responsible drug usage offers an integrated strategy for managing the antibiotic resistance challenge.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (G.C.); (E.D.)
| | - Carolina Pérez Reyes
- Department of Biochemistry, Microbiology, Cell Biology and Genetics, Instituto Universitario de Bio-Orgánica “Antonio González”, University of La Laguna, 38206 San Cristobal de La Laguna, Spain;
| | - Davide Laurenti
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (D.L.); (C.C.); (R.M.); (D.D.R.)
| | - Carmen Chiaradia
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (D.L.); (C.C.); (R.M.); (D.D.R.)
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (G.C.); (E.D.)
| | - Roberto Mattioli
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (D.L.); (C.C.); (R.M.); (D.D.R.)
| | - Daniel Di Risola
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (D.L.); (C.C.); (R.M.); (D.D.R.)
| | | | - Antonio Francioso
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (G.C.); (E.D.)
| |
Collapse
|
20
|
Yalçın MS, Özdemir S, Prokopiuk V, Virych P, Onishchenko A, Tollu G, Pavlenko V, Kutsevol N, Dizge N, Tkachenko A, Ocakoglu K. Toxicity, Antibacterial, Antioxidant, Antidiabetic, and DNA Cleavage Effects of Dextran-Graft-Polyacrylamide/Zinc Oxide Nanosystems. Curr Microbiol 2024; 81:437. [PMID: 39487865 DOI: 10.1007/s00284-024-03953-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/12/2024] [Indexed: 11/04/2024]
Abstract
Synthesis of metal oxide nanoparticles-polymer nanocomposites is an emerging strategy in nanotechnology to improve targeted delivery and reduce the toxicity of nanoparticles. In this study, we report biological effects of previously described hybrid nanocomposites containing dextran-graft-polyacrylamide/zinc oxide nanoparticles (D-PAA/ZnO NPs) prepared from zinc sulfate (D-PAA/ZnONPs(SO42-)) and zinc acetate (D-PAA/ZnONPs(-OAc)) focusing primarily on their antimicrobial activity. D-PAA/ZnONPs(SO42-) and D-PAA/ZnONPs(-OAc) nanosystems were tested in a complex way to assess their antioxidant activity (DPPH assay), antidiabetic potential (α-amylase inhibition), DNA cleavage activity, antimicrobial, and antibiofilm activity. In addition, the toxicity of D-PAA/ZnONPs(SO42-) and D-PAA/ZnONPs(-OAc) nanosystems against primary murine splenocytes was tested using MTT assay. The studied nanosystems inhibited E.coli growth. For all the investigated strains, minimum inhibitory concentrations (MICs) of D-PAA/ZnONPs(SO42-) and D-PAA/ZnONPs(-OAc) were in the range of 8 mg/L-128 mg/L and 16 mg/L-128 mg/L, respectively. The nanocomposites demonstrated effective antibiofilm properties as 94.27% and 86.43%. The compounds showed good antioxidant, anti-α-amylase, and DNA cleavage activities. D-PAA/ZnONPs(SO42-) and D-PAA/ZnONPs(-OAc) nanosystems reduced cell viability and promoted cell death of primary murine spleen cells at concentrations higher than those that proved to be antibacterial indicating the presence of therapeutic window. D-PAA/ZnONPs(SO42-) and D-PAA/ZnONPs(-OAc) nanosystems show antioxidant, antidiabetic, DNA cleavage, antimicrobial, and antibiofilm activity against the background of good biocompatibility suggesting the presence of therapeutic potential, which should be further investigated in vivo.
Collapse
Affiliation(s)
- M Serkan Yalçın
- Department of Chemistry and Chemical Processing Technologies, Technical Science Vocational School, Mersin University, 33343, Mersin, Turkey
| | - Sadin Özdemir
- Food Processing Programme, Technical Science Vocational School, Mersin University, TR-33343, Yenisehir, Mersin, Turkey
| | - Volodymyr Prokopiuk
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, 61022, Ukraine
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, Kharkiv, 61015, Ukraine
| | - Pavlo Virych
- Department of Chemistry, Taras Shevchenko National University of Kyiv, Kyiv, 01601, Ukraine
| | - Anatolii Onishchenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, Kharkiv, 61015, Ukraine
| | - Gülşah Tollu
- Laboratory and Veterinary Health, Technical Science Vocational School, Mersin University, Yenisehir, TR-33343, Mersin, Turkey
| | - Vadim Pavlenko
- Department of Chemistry, Taras Shevchenko National University of Kyiv, Kyiv, 01601, Ukraine
| | - Nataliya Kutsevol
- Department of Chemistry, Taras Shevchenko National University of Kyiv, Kyiv, 01601, Ukraine
| | - Nadir Dizge
- Department of Environmental Engineering, Mersin University, Yenişehir, 33343, Mersin, Turkey
| | - Anton Tkachenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, Kharkiv, 61015, Ukraine.
| | - Kasim Ocakoglu
- Department of Eng. Fundamental Sciences, Faculty of Engineering, Tarsus University, 33400, Tarsus, Turkey
| |
Collapse
|
21
|
de Farias BO, Saggioro EM, Montenegro KS, Magaldi M, Santos HSO, Gonçalves-Brito AS, Pimenta RL, Ferreira RG, Spisso BF, Pereira MU, Bianco K, Clementino MM. Metagenomic insights into plasmid-mediated antimicrobial resistance in poultry slaughterhouse wastewater: antibiotics occurrence and genetic markers. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:60880-60894. [PMID: 39395082 DOI: 10.1007/s11356-024-35287-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Slaughterhouse wastewater represents important convergence and concentration points for antimicrobial residues, bacteria, and antibiotic resistance genes (ARG), which can promote antimicrobial resistance propagation in different environmental compartments. This study reports the assessment of the metaplasmidome-associated resistome in poultry slaughterhouse wastewater treated by biological processes, employing metagenomic sequencing. Antimicrobial residues from a wastewater treatment plant (WWTP) that treats poultry slaughterhouse influents and effluents were investigated through high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS). Residues from the macrolide, sulfonamide, and fluoroquinolone classes were detected, the latter two persisting after the wastewater treatment. The genetic markers 16S rRNA rrs (bacterial community) and uidA (Escherichia coli) were investigated by RT-qPCR and the sul1 and int1 genes by qPCR. After treatment, the 16S rRNA rrs, uidA, sul1, and int1 markers exhibited reductions of 0.67, 1.07, 1.28, and 0.79 genes copies, respectively, with no statistical significance (p > 0.05). The plasmidome-focused metagenomics sequences (MiSeq platform (Illumina®)) revealed more than 100 ARG in the WWTP influent, which can potentially confer resistance to 14 pharmacological classes relevant in the human and veterinary clinical contexts, in which the qnr gene (resistance to fluoroquinolones) was the most prevalent. Only 7.8% of ARG were reduced after wastewater treatment, and the remaining 92.2% were associated with an increase in the prevalence of ARG linked to multidrug efflux pumps, substrate-specific for certain classes of antibiotics, or broad resistance to multiple medications. These data demonstrate that wastewater from poultry slaughterhouses plays a crucial role as an ARG reservoir and in the spread of AMR into the environment.
Collapse
Affiliation(s)
- Beatriz Oliveira de Farias
- Programa de Pós-Graduação Em Saúde Pública E Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Enrico Mendes Saggioro
- Programa de Pós-Graduação Em Saúde Pública E Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil.
- Laboratório de Avaliação E Promoção da Saúde Ambiental, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil.
| | - Kaylanne S Montenegro
- Programa de Pós-Graduação Em Saúde Pública E Meio Ambiente, Escola Nacional de Saúde Pública, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Mariana Magaldi
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Hugo Sérgio Oliveira Santos
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Andressa Silva Gonçalves-Brito
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Ramon Loureiro Pimenta
- Instituto de Veterinária, Universidade Federal Rural Do Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ, Brazil
| | - Rosana Gomes Ferreira
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Bernardete Ferraz Spisso
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Mararlene Ulberg Pereira
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Kayo Bianco
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Maysa Mandetta Clementino
- Instituto Nacional de Controle de Qualidade Em Saúde, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
22
|
Brdová D, Ruml T, Viktorová J. Mechanism of staphylococcal resistance to clinically relevant antibiotics. Drug Resist Updat 2024; 77:101147. [PMID: 39236354 DOI: 10.1016/j.drup.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Staphylococcus aureus, a notorious pathogen with versatile virulence, poses a significant challenge to current antibiotic treatments due to its ability to develop resistance mechanisms against a variety of clinically relevant antibiotics. In this comprehensive review, we carefully dissect the resistance mechanisms employed by S. aureus against various antibiotics commonly used in clinical settings. The article navigates through intricate molecular pathways, elucidating the mechanisms by which S. aureus evades the therapeutic efficacy of antibiotics, such as β-lactams, vancomycin, daptomycin, linezolid, etc. Each antibiotic is scrutinised for its mechanism of action, impact on bacterial physiology, and the corresponding resistance strategies adopted by S. aureus. By synthesising the knowledge surrounding these resistance mechanisms, this review aims to serve as a comprehensive resource that provides a foundation for the development of innovative therapeutic strategies and alternative treatments for S. aureus infections. Understanding the evolving landscape of antibiotic resistance is imperative for devising effective countermeasures in the battle against this formidable pathogen.
Collapse
Affiliation(s)
- Daniela Brdová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| |
Collapse
|
23
|
Khan RT, Sharma V, Khan SS, Rasool S. Prevention and potential remedies for antibiotic resistance: current research and future prospects. Front Microbiol 2024; 15:1455759. [PMID: 39421555 PMCID: PMC11484029 DOI: 10.3389/fmicb.2024.1455759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The increasing threat of antibiotic resistance and shrinking treatment options for infections have pushed mankind into a difficult position. The looming threat of the return of the pre-antibiotic era has caused a sense of urgency to protect and conserve the potency of antibiotic therapy. One of the perverse effects of antibiotic resistance is the dissemination of its causative agents from non-clinically important strains to clinically important strains and vice versa. The popular saying "Prevention is better than cure" is appropriate for tackling antibiotic resistance. On the one hand, new and effective antibiotics are required; on the other hand, better measures for the use of antibiotics, along with increased awareness in the general public related to antibiotic use, are essential. Awareness, especially of appropriate antibiotic use, antibiotic resistance, its dissemination, and potential threats, can help greatly in controlling the use and abuse of antibiotics, and the containment of antibiotic resistance. Antibiotic drugs' effectiveness can be enhanced by producing novel antibiotic analogs or adding adjuvants to current antibiotics. Combinatorial therapy of antibiotics has proven successful in treating multidrug-resistant (MDR) bacterial infections. This review aims to highlight the current global situation of antibiotic resistance and discuss the methods used to monitor, prevent, inhibit, or reverse bacterial resistance mechanisms in the fight against antibiotic resistance.
Collapse
Affiliation(s)
| | | | | | - Shafaq Rasool
- Molecular Biology Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| |
Collapse
|
24
|
Khan NA, Alvi A, Alqassim S, Akbar N, Khatoon B, Kawish M, Faizi S, Shah MR, Alawfi BS, Siddiqui R. Nanomedicine: Patuletin-conjugated with zinc oxide exhibit potent effects against Gram-negative and Gram-positive bacterial pathogens. Biometals 2024; 37:1113-1125. [PMID: 38705945 DOI: 10.1007/s10534-024-00595-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 03/02/2024] [Indexed: 05/07/2024]
Abstract
With the emergence of drug-resistance, there is a need for novel anti-bacterials or to enhance the efficacy of existing drugs. In this study, Patuletin (PA), a flavanoid was loaded onto Gallic acid modified Zinc oxide nanoparticles (PA-GA-ZnO), and evaluated for antibacterial properties against Gram-positive (Bacillus cereus and Streptococcus pneumoniae) and Gram-negative (Samonella enterica and Escherichia coli) bacteria. Characterization of PA, GA-ZnO and PA-GA-ZnO' nanoparticles was accomplished utilizing fourier-transform infrared spectroscopy, efficiency of drug entrapment, polydispersity index, zeta potential, size, and surface morphology analysis through atomic force microscopy. Using bactericidal assays, the results revealed that ZnO conjugation displayed remarkable effects and enhanced Patuletin's effects against both Gram-positive and Gram-negative bacteria, with the minimum inhibitory concentration observed at micromolar concentrations. Cytopathogenicity assays exhibited that the drug-nanoconjugates reduced bacterial-mediated human cell death with minimal side effects to human cells. When tested alone, drug-nanoconjugates tested in this study showed limited toxic effects against human cells in vitro. These are promising findings, but future work is needed to understand the molecular mechanisms of effects of drug-nanoconjugates against bacterial pathogens, in addition to in vivo testing to determine their translational value. This study suggests that Patuletin-loaded nano-formulation (PA-GA-ZnO) may be implicated in a multi-target mechanism that affects both Gram-positive and Gram-negative pathogen cell structures, however this needs to be ascertained in future work.
Collapse
Affiliation(s)
- Naveed Ahmed Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey.
| | - Adeelah Alvi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE
| | - Saif Alqassim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE.
| | - Noor Akbar
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Bushra Khatoon
- International Center for Chemical and Biological Sciences, H.E.J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Kawish
- International Center for Chemical and Biological Sciences, H.E.J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Shaheen Faizi
- International Center for Chemical and Biological Sciences, H.E.J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Raza Shah
- International Center for Chemical and Biological Sciences, H.E.J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Bader S Alawfi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, 42353, Madinah, Saudi Arabia
| | - Ruqaiyyah Siddiqui
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, , Edinburgh, EH14 4AS, UK
| |
Collapse
|
25
|
Jamwal V, Palmo T, Singh K. Understanding the mechanisms of antimicrobial resistance and potential therapeutic approaches against the Gram-negative pathogen Acinetobacter baumannii. RSC Med Chem 2024; 15:d4md00449c. [PMID: 39386059 PMCID: PMC11457259 DOI: 10.1039/d4md00449c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Globally, the emergence of anti-microbial resistance in pathogens has become a serious threat to human health and well-being. Infections caused by drug-resistant microorganisms in hospitals are associated with increased morbidity, mortality, and healthcare costs. Acinetobacter baumannii is a Gram-negative bacterium belonging to the ESKAPE group and is widely associated with nosocomial infections. It persists in hospitals and survives antibiotic treatment, prompting acute infections such as urinary tract infections, pneumonia, bacteremia, meningitis, and wound-related infections. An innovation void in drug discovery and the lack of new therapeutic measures against A. baumannii continue to afflict infection control against the rising drug-resistant cases. The emergence of drug-resistant A. baumannii strains has also led to the incessant collapse of newly discovered antibiotics. Therefore exploring novel strategies is requisite to give impetus to A. baumannii drug discovery. The present review discusses the bacterial research community's efforts in the field of A. baumannii, focusing on the strategies adapted to identify potent scaffolds and novel targets to bolster and diversify the chemical space available for drug discovery. Firstly, we have discussed existing chemotherapy and various anti-microbial resistance mechanisms in A. baumannii bacterial strains. Next, we elaborate on multidisciplinary approaches and strategies that may be the way forward to combat the current menace caused by the drug-resistant A. baumannii strains. The review highlights the recent advances in drug discovery, including combinational therapy, high-throughput screening, drug repurposing, nanotechnology, and anti-microbial peptides, which are imperative tools to fight bacterial pathogens in the future.
Collapse
Affiliation(s)
- Vishwani Jamwal
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Tashi Palmo
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| |
Collapse
|
26
|
Punchihewage-Don AJ, Ranaweera PN, Parveen S. Defense mechanisms of Salmonella against antibiotics: a review. FRONTIERS IN ANTIBIOTICS 2024; 3:1448796. [PMID: 39816264 PMCID: PMC11731628 DOI: 10.3389/frabi.2024.1448796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/26/2024] [Indexed: 01/18/2025]
Abstract
Salmonella is a foodborne pathogenic bacterium that causes salmonellosis worldwide. Also, Salmonella is considered a serious problem for food safety and public health. Several antimicrobial classes including aminoglycosides, tetracyclines, phenols, and β-Lactams are used to treat Salmonella infections. Antibiotics have been prescribed for decades to treat infections caused by bacteria in human and animal healthcare. However, intensive use of antibiotics resulted in antibiotic resistance (AR) among several foodborne bacteria including Salmonella. Furthermore, multi-drug resistance (MDR) of Salmonella has increased dramatically. In addition to MDR Salmonella, extensively drug resistant (XDR) as well as pan drug resistant (PDR) Salmonella were reported globally. Therefore, increasing AR is becoming a serious universal public health crisis. Salmonella developed many mechanisms to ensure its survival against antimicrobials. The most prominent defense mechanisms against these antibiotics include enzymatic inactivation, expelling drugs from the cell through efflux pumps, altering the structure of drugs, and changing or protecting the targets of drugs. Additionally, the formation of biofilms and plasmid-mediated AR by Salmonella, enhancing its resistance to various antibiotics, making it a challenging pathogen in both healthcare and food industry settings. This review focuses exclusively on providing a detailed overview of the mechanisms of AR in Salmonella.
Collapse
Affiliation(s)
| | | | - Salina Parveen
- Department of Agriculture, Food, and Resource Sciences, University of Maryland Eastern Shore, Princess Anne, MD, United States
| |
Collapse
|
27
|
Onyeka LO, Adesiyun AA, Ismail A, Allam M, Keddy KH, Thompson PN. Evidence for Horizontal Transmission and Recirculation of Shiga Toxin-Producing Escherichia coli in the Beef Production Chain in South Africa Using Whole Genome Sequencing. Pathogens 2024; 13:732. [PMID: 39338923 PMCID: PMC11434950 DOI: 10.3390/pathogens13090732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
We used whole genome sequencing (WGS) as an epidemiologic surveillance tool to elucidate the transmission dynamics of Shiga toxin-producing Escherichia coli (STEC) strains along the beef production chain in South Africa. Isolates were obtained from a cattle farm, abattoirs and retail outlets. Isolates were analysed using WGS on a MiSeq platform (Illumina, San Diego, CA, USA) and phylogenetic analysis was carried out. Of the 85 isolates, 39% (33) carried the stx gene and 61% (52) had lost the stx gene. The prevalence of stx subtypes was as follows; stx1a 55% (18/33), stx1b 52% (17/33), stx2a 55% (18/33), stx2b 27% (9/33), stx2dB 30% (10/33) and stx2d1A 15% (5/33). Thirty-five different serogenotypes were detected, of which 65% (56) were flagellar H-antigens and 34% (29) were both O-antigens and flagellar H-antigens. We identified 50 different sequence types (STs), and only nine of the isolates were assigned to three different clonal complexes. Core genome phylogenetic analysis revealed genetic relatedness, and isolates clustered mainly according to their STs and serogenotypes regardless of stx subtypes. This study provides evidence of horizontal transmission and recirculation of STEC strains in Gauteng province and demonstrates that every stage of the beef production chain plays a significant role in STEC entry into the food chain.
Collapse
Affiliation(s)
- Libby Obumneke Onyeka
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa;
- Department of Veterinary Public Health and Preventive Medicine, College of Veterinary Medicine, Michael Okpara University of Agriculture, Umudike 440101, Abia State, Nigeria
| | - Abiodun A. Adesiyun
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa;
- Department of Basic Veterinary Sciences, School of Veterinary Medicine, Faculty of Medical Sciences, University of the West Indies, St. Augustine 999183, Trinidad and Tobago
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases a Division of the National Health Laboratory Service, Johannesburg 2192, South Africa; (A.I.); (M.A.)
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou 0950, South Africa
- Institute for Water and Wastewater Technology, Durban University of Technology, Durban 4000, South Africa
| | - Mushal Allam
- Sequencing Core Facility, National Institute for Communicable Diseases a Division of the National Health Laboratory Service, Johannesburg 2192, South Africa; (A.I.); (M.A.)
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Karen H. Keddy
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa;
| | - Peter N. Thompson
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa;
| |
Collapse
|
28
|
Akanksha, Mehra S. Conserved Evolutionary Trajectory Can Be Perturbed to Prevent Resistance Evolution under Norfloxacin Pressure by Forcing Mycobacterium smegmatis on Alternate Evolutionary Paths. ACS Infect Dis 2024; 10:2623-2636. [PMID: 38959403 DOI: 10.1021/acsinfecdis.3c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Antibiotic resistance is a pressing health issue, with the emergence of resistance in bacteria outcompeting the discovery of novel drug candidates. While many studies have used Adaptive Laboratory Evolution (ALE) to understand the determinants of resistance, the influence of the drug dosing profile on the evolutionary trajectory remains understudied. In this study, we employed ALE on Mycobacterium smegmatis exposed to various concentrations of Norfloxacin using both cyclic constant and stepwise increasing drug dosages to examine their impact on the resistance mechanisms selected. Mutations in an efflux pump regulator, LfrR, were found in all of the evolved populations irrespective of the drug profile and population bottleneck, indicating a conserved efflux-based resistance mechanism. This mutation appeared early in the evolutionary trajectory, providing low-level resistance when present alone, with a further increase in resistance resulting from successive accumulation of other mutations. Notably, drug target mutations, similar to those observed in clinical isolates, were only seen above a threshold of greater than 4× the minimum inhibitory concentration (MIC). A combination of three mutations in the genes, lfrR, MSMEG_1959, and MSMEG_5045, was conserved across multiple lineages, leading to high-level resistance and preceding the appearance of drug target mutations. Interestingly, in populations evolved from parental strains lacking the lfrA efflux pump, the primary target of the lfrR regulator, no lfrR gene mutations are selected. Furthermore, evolutional trajectories originating from the ΔlfrA strain displayed early arrest in some lineages and the absence of target gene mutations in those that evolved, albeit delayed. Thus, blocking or inhibiting the expression of efflux pumps can arrest or delay the fixation of drug target mutations, potentially limiting the maximum attainable resistance levels.
Collapse
Affiliation(s)
- Akanksha
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Sarika Mehra
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| |
Collapse
|
29
|
Norouzalinia F, Asadpour L, Mokhtary M. Anti-microbial, anti-biofilm, and efflux pump inhibitory effects of ellagic acid-bonded magnetic nanoparticles against Escherichia coli isolates. Int Microbiol 2024:10.1007/s10123-024-00560-4. [PMID: 39105888 DOI: 10.1007/s10123-024-00560-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/06/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
The spread of microbial resistance is a threat to public health. In this study, the anti-microbial, anti-biofilm, and efflux pump inhibitory effects of ellagic acid-loaded magnetic nanoparticles (Fe3O4NPs@EA) against beta-lactamase producing Escherichia coli isolates have been investigated. The effects of Fe3O4 NPs@EA on the growth inhibition of E. coli isolates were determined by disc diffusion method and determining the minimum inhibitory concentration was done using broth micro-dilution method. The anti-biofilm effect of nanoparticles was investigated using the microplate method. The efflux pump inhibitory effect of nanoparticles was investigated using cart-wheel method and by investigating the effect of nanoparticles on acrB and tolC genes expression levels. Fe3O4 NPs@EA showed anti-bacterial effects against test bacteria, and the MIC of these nanoparticles varied from 0.19 to 1.56 mg/mL. These nanoparticles caused a 43-62% reduction in biofilm formation of test bacteria compared to control. Furthermore, efflux pump inhibitory effect of these nanoparticles was confirmed at a concentration of 1/8 MIC, and the expression of acrB and tolC genes decreased in bacteria treated with 1/4 MIC Fe3O4 NPs@EA. According to the results, the use of nanoparticles containing ellagic acid can provide a basis for the development of new treatments against drug-resistant E. coli. This substance may improve the concentration of antibiotics in the bacterial cell and increase their effectiveness by inhibiting the efflux in E. coli isolates.
Collapse
Affiliation(s)
| | - Leila Asadpour
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Masoud Mokhtary
- Department of Chemistry, Rasht Branch, Islamic Azad University, Rasht, Iran
| |
Collapse
|
30
|
Wang WX, Wu JZ, Zhang BL, Yu JY, Han LM, Lu XL, Li H, Fu SY, Ren YY, Dong H, Xu Y, Wang GT, Gao JH, Wang C, Chen XZ, Liu DX, Huang Y, Yu JH, Wang SW, Yang YF, Chen W. Phage therapy combats pan drug-resistant Acinetobacter baumannii infection safely and efficiently. Int J Antimicrob Agents 2024; 64:107220. [PMID: 38810939 DOI: 10.1016/j.ijantimicag.2024.107220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 04/20/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Phage therapy offers a promising approach to combat the growing threat of antimicrobial resistance. Yet, key questions remain regarding dosage, administration routes, combination therapy, and the causes of therapeutic failure. In this study, we focused on a novel lytic phage, ФAb4B, which specifically targeted the Acinetobacter baumannii strains with KL160 capsular polysaccharide, including the pan-drug resistant A. baumannii YQ4. ФAb4B exhibited the ability to effectively inhibit biofilm formation and eradicate mature biofilms independently of dosage. Additionally, it demonstrated a wide spectrum of antibiotic-phage synergy and did not show any cytotoxic or haemolytic effects. Continuous phage injections, both intraperitoneally and intravenously over 7 d, showed no acute toxicity in vivo. Importantly, phage therapy significantly improved neutrophil counts, outperforming ciprofloxacin. However, excessive phage injections suppressed neutrophil levels. The combinatorial treatment of phage-ciprofloxacin rescued 91% of the mice, a superior outcome compared to phage alone (67%). The efficacy of the combinatorial treatment was independent of phage dosage. Notably, prophylactic administration of the combinatorial regimen provided no protection, but even when combined with a delayed therapeutic regimen, it saved all the mice. Bacterial resistance to the phage was not a contributing factor to treatment failure. Our preclinical study systematically describes the lytic phage's effectiveness in both in vitro and in vivo settings, filling in crucial details about phage treatment against bacteriemia caused by A. baumannii, which will provide a robust foundation for the future of phage therapy.
Collapse
Affiliation(s)
- Wei-Xiao Wang
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jia-Zhen Wu
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bai-Ling Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiao-Yang Yu
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Li-Mei Han
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao-Liang Lu
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Hui Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shi-Yong Fu
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Yao Ren
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Dong
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Xu
- Department of Geriatric Medicine, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Gong-Ting Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Jing-Han Gao
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Chun Wang
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiu-Zhen Chen
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Du-Xian Liu
- Department of pathology, the Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Huang
- Department of Infection Control and Management, the Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin-Hong Yu
- Department of Clinical Laboratory, the Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shi-Wei Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Yong-Feng Yang
- The Clinical Infectious Disease Center of Nanjing, Nanjing, China.
| | - Wei Chen
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
31
|
Urra G, Valdés-Muñoz E, Suardiaz R, Hernández-Rodríguez EW, Palma JM, Ríos-Rozas SE, Flores-Morales CA, Alegría-Arcos M, Yáñez O, Morales-Quintana L, D’Afonseca V, Bustos D. From Proteome to Potential Drugs: Integration of Subtractive Proteomics and Ensemble Docking for Drug Repurposing against Pseudomonas aeruginosa RND Superfamily Proteins. Int J Mol Sci 2024; 25:8027. [PMID: 39125594 PMCID: PMC11312079 DOI: 10.3390/ijms25158027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) poses a significant threat as a nosocomial pathogen due to its robust resistance mechanisms and virulence factors. This study integrates subtractive proteomics and ensemble docking to identify and characterize essential proteins in P. aeruginosa, aiming to discover therapeutic targets and repurpose commercial existing drugs. Using subtractive proteomics, we refined the dataset to discard redundant proteins and minimize potential cross-interactions with human proteins and the microbiome proteins. We identified 12 key proteins, including a histidine kinase and members of the RND efflux pump family, known for their roles in antibiotic resistance, virulence, and antigenicity. Predictive modeling of the three-dimensional structures of these RND proteins and subsequent molecular ensemble-docking simulations led to the identification of MK-3207, R-428, and Suramin as promising inhibitor candidates. These compounds demonstrated high binding affinities and effective inhibition across multiple metrics. Further refinement using non-covalent interaction index methods provided deeper insights into the electronic effects in protein-ligand interactions, with Suramin exhibiting superior binding energies, suggesting its broad-spectrum inhibitory potential. Our findings confirm the critical role of RND efflux pumps in antibiotic resistance and suggest that MK-3207, R-428, and Suramin could be effectively repurposed to target these proteins. This approach highlights the potential of drug repurposing as a viable strategy to combat P. aeruginosa infections.
Collapse
Affiliation(s)
- Gabriela Urra
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| | - Elizabeth Valdés-Muñoz
- Doctorado en Biotecnología Traslacional, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca 3480094, Chile;
| | - Reynier Suardiaz
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Erix W. Hernández-Rodríguez
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
- Unidad de Bioinformática Clínica, Centro Oncológico, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile
| | - Jonathan M. Palma
- Facultad de Ingeniería, Universidad de Talca, Curicó 3344158, Chile;
| | - Sofía E. Ríos-Rozas
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| | | | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile; (M.A.-A.); (O.Y.)
| | - Osvaldo Yáñez
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile; (M.A.-A.); (O.Y.)
| | - Luis Morales-Quintana
- Multidisciplinary Agroindustry Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Cinco Pte. N° 1670, Talca 3467987, Chile;
| | - Vívian D’Afonseca
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad Católica del Maule, Ave. San Miguel 3605, Talca 3466706, Chile
| | - Daniel Bustos
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile; (G.U.); (E.W.H.-R.); (S.E.R.-R.)
| |
Collapse
|
32
|
Deshpande A, Likhar R, Khan T, Omri A. Decoding drug resistance in Mycobacterium tuberculosis complex: genetic insights and future challenges. Expert Rev Anti Infect Ther 2024; 22:511-527. [PMID: 39219506 DOI: 10.1080/14787210.2024.2400536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/02/2024] [Accepted: 08/31/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Tuberculosis (TB), particularly its drug-resistant forms (MDR-TB and XDR-TB), continues to pose a significant global health challenge. Despite advances in treatment and diagnosis, the evolving nature of drug resistance in Mycobacterium tuberculosis (MTB) complicates TB eradication efforts. This review delves into the complexities of anti-TB drug resistance, its mechanisms, and implications on healthcare strategies globally. AREAS COVERED We explore the genetic underpinnings of resistance to both first-line and second-line anti-TB drugs, highlighting the role of mutations in key genes. The discussion extends to advanced diagnostic techniques, such as Whole-Genome Sequencing (WGS), CRISPR-based diagnostics and their impact on identifying and managing drug-resistant TB. Additionally, we discuss artificial intelligence applications, current treatment strategies, challenges in managing MDR-TB and XDR-TB, and the global disparities in TB treatment and control, translating to different therapeutic outcomes and have the potential to revolutionize our understanding and management of drug-resistant tuberculosis. EXPERT OPINION The current landscape of anti-TB drug resistance demands an integrated approach combining advanced diagnostics, novel therapeutic strategies, and global collaborative efforts. Future research should focus on understanding polygenic resistance and developing personalized medicine approaches. Policymakers must prioritize equitable access to diagnosis and treatment, enhancing TB control strategies, and support ongoing research and augmented government funding to address this critical public health issue effectively.
Collapse
Affiliation(s)
- Amey Deshpande
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, India
| | - Rupali Likhar
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Chemistry, LSHGCT's Gahlot Institute of Pharmacy, Navi Mumbai, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
33
|
Wu J, Liu L, Wang J, Wang Y, Li X, Wang X, Jiang S, Li W, Zhang J, Zhang X. Transcriptomic analysis of induced resistance to polymyxin in carbapenem-resistant Enterobacter cloacae complex isolate carrying mcr-9. J Glob Antimicrob Resist 2024; 37:225-232. [PMID: 38750896 DOI: 10.1016/j.jgar.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 03/17/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVES Polymyxins are currently the last-resort treatment against multi-drug resistant Gram-negative bacterial infections, but plasmid-mediated mobile polymyxin resistance genes (mcr) threaten its efficacy, especially in carbapenem-resistant Enterobacter cloacae complex (CRECC). The objective of this study was to provide insights into the mechanism of polymyxin-induced bacterial resistance and the effect of overexpression of mcr-9. METHODS The clinical strain CRECC414 carrying the mcr-9 gene was treated with a gradient concentration of polymyxin. Subsequently, the broth microdilution was used to determine the minimum inhibitory concentration (MIC) and RT-qPCR was utilized to assess mcr-9 expression. Transcriptome sequencing and whole genome sequencing (WGS) was utilized to identify alterations in strains resulting from increased polymyxin resistance, and significant transcriptomic differences were analysed alongside a comprehensive examination of metabolic networks at the genomic level. RESULTS Polymyxin treatment induced the upregulation of mcr-9 expression and significantly elevated the MIC of the strain. Furthermore, the WGS and transcriptomic results revealed a remarkable up-regulation of arnBCADTEF gene cassette, indicating that the Arn/PhoPQ system-mediated L-Ara4N modification is the preferred mechanism for achieving high levels of resistance. Additionally, significant alterations in bacterial gene expression were observed with regards to multidrug efflux pumps, oxidative stress and repair mechanisms, cell membrane biosynthesis, as well as carbohydrate metabolic pathways. CONCLUSION Polymyxin greatly disrupts the transcription of vital cellular pathways. A complete PhoPQ two-component system is a prerequisite for polymyxin resistance of Enterobacter cloacae, even though mcr-9 is highly expressed. These findings provide novel and important information for further investigation of polymyxin resistance of CRECC.
Collapse
Affiliation(s)
- Jiming Wu
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Longjin Liu
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Jianmin Wang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Wang
- Department of Microbiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Xinhui Li
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Wang
- Department of Microbiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shan Jiang
- Department of Pathogenic Biology, Basic Medicine of Jiamusi University, Jiamusi, China
| | - Wengang Li
- Department of Pathogenic Biology, Basic Medicine of Jiamusi University, Jiamusi, China
| | - Jisheng Zhang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaoli Zhang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
34
|
Carvalho R, Tapia JH, Minsavage GV, Jones JB, Paret ML. Elucidating the Mode of Action of Hybrid Nanoparticles of Cu/Zn Against Copper-Tolerant Xanthomonas euvesicatoria. PHYTOPATHOLOGY 2024; 114:1206-1214. [PMID: 38302452 DOI: 10.1094/phyto-09-23-0339-r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
The widespread presence of tolerance to copper in Xanthomonas species has resulted in the need to develop alternative approaches to control plant diseases caused by xanthomonads. In recent years, nanotechnological approaches have resulted in the identification of novel materials to control plant pathogens. With many metal-based nanomaterials having shown promise for disease control, an important question relates to the mode of action of these new materials. In this study, we used several approaches, such as scanning electron microscopy, propidium monoazide quantitative polymerase chain reaction, epifluorescence microscopy, and RNA sequencing to elucidate the mode of action of a Cu/Zn hybrid nanoparticle against copper-tolerant strains of Xanthomonas euvesicatoria. We demonstrate that Cu/Zn did not activate copper resistance genes (i.e., copA and copB) in the copper-tolerant bacterium but functioned by disrupting the bacterial cell structure and perturbing important biological processes such as cell respiration and chemical homeostasis.
Collapse
Affiliation(s)
- Renato Carvalho
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
| | - Jose H Tapia
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
| | - Gerald V Minsavage
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
| | - Jeffrey B Jones
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
| | - Mathews L Paret
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611
- North Florida Research and Education Center, University of Florida, Quincy, FL 32251
| |
Collapse
|
35
|
Motta H, Reuwsaat JCV, Lopes FC, Viezzer G, Volpato FCZ, Barth AL, de Tarso Roth Dalcin P, Staats CC, Vainstein MH, Kmetzsch L. Comparative microbiome analysis in cystic fibrosis and non-cystic fibrosis bronchiectasis. Respir Res 2024; 25:211. [PMID: 38762736 PMCID: PMC11102160 DOI: 10.1186/s12931-024-02835-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND Bronchiectasis is a condition characterized by abnormal and irreversible bronchial dilation resulting from lung tissue damage and can be categorized into two main groups: cystic fibrosis (CF) and non-CF bronchiectasis (NCFB). Both diseases are marked by recurrent infections, inflammatory exacerbations, and lung damage. Given that infections are the primary drivers of disease progression, characterization of the respiratory microbiome can shed light on compositional alterations and susceptibility to antimicrobial drugs in these cases compared to healthy individuals. METHODS To assess the microbiota in the two studied diseases, 35 subjects were recruited, comprising 10 NCFB and 13 CF patients and 12 healthy individuals. Nasopharyngeal swabs and induced sputum were collected, and total DNA was extracted. The DNA was then sequenced by the shotgun method and evaluated using the SqueezeMeta pipeline and R. RESULTS We observed reduced species diversity in both disease cohorts, along with distinct microbial compositions and profiles of antimicrobial resistance genes, compared to healthy individuals. The nasopharynx exhibited a consistent microbiota composition across all cohorts. Enrichment of members of the Burkholderiaceae family and an increased Firmicutes/Bacteroidetes ratio in the CF cohort emerged as key distinguishing factors compared to NCFB group. Staphylococcus aureus and Prevotella shahii also presented differential abundance in the CF and NCFB cohorts, respectively, in the lower respiratory tract. Considering antimicrobial resistance, a high number of genes related to antibiotic efflux were detected in both disease groups, which correlated with the patient's clinical data. CONCLUSIONS Bronchiectasis is associated with reduced microbial diversity and a shift in microbial and resistome composition compared to healthy subjects. Despite some similarities, CF and NCFB present significant differences in microbiome composition and antimicrobial resistance profiles, suggesting the need for customized management strategies for each disease.
Collapse
Affiliation(s)
- Heryk Motta
- Laboratório de Biologia Molecular de Patógenos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Júlia Catarina Vieira Reuwsaat
- Laboratório de Biologia Molecular de Patógenos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Cortez Lopes
- Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Graciele Viezzer
- Serviço de Pneumologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fabiana Caroline Zempulski Volpato
- Laboratório de Pesquisa em Resistência Bacteriana, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Afonso Luís Barth
- Laboratório de Pesquisa em Resistência Bacteriana, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Paulo de Tarso Roth Dalcin
- Serviço de Pneumologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Charley Christian Staats
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marilene Henning Vainstein
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Microrganismos de Importância Médica e Biotecnológica, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lívia Kmetzsch
- Laboratório de Biologia Molecular de Patógenos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
36
|
Dan MO, Tǎlǎpan D. Friends or foes? Novel antimicrobials tackling MDR/XDR Gram-negative bacteria: a systematic review. Front Microbiol 2024; 15:1385475. [PMID: 38800756 PMCID: PMC11116650 DOI: 10.3389/fmicb.2024.1385475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
Gram-negative bacteria have been one of the most studied classes in the field of microbiology, especially in the context of globally alarming antimicrobial resistance levels to these pathogens over the course of the past decades. With high numbers of these microorganisms being described as multidrug-resistant (MDR), or even extended-drug-resistant (XDR) bacteria, specialists in the field have been struggling to keep up with higher prevalence of difficult-to-treat infections caused by such superbugs. The FDA approval of novel antimicrobials, such as cefiderocol (FDC), ceftolozane/tazobactam (C/T), ceftazidime/avibactam (CZA), imipenem/relebactam (IMR), sulbactam/durlobactam (SUL-DUR) and phase 3 clinical trials' results of aztreonam/avibactam (ATM-AVI) has proven that, while all these substances provide encouraging efficacy rates, antibiotic resistance keeps up with the pace of drug development. Microorganisms have developed more extensive mechanisms of resistance in order to target the threat posed by these novel antimicrobials, thus equiring researchers to be on a constant lookout for other potential drug candidates and molecule development. However, these strategies require a proper understanding of bacterial resistance mechanisms to gain a comprehensive outlook on the issue. The present review aims to highlight these six antibiotic agents, which have brought hope to clinicians during the past decade, discussing general properties of these substances, as well as mechanisms and patterns of resistance, while also providing a short overview on further directions in the field. Systematic review registration https://www.crd.york.ac.uk/prospero/#searchadvanced, Identifier CRD42024505832.
Collapse
Affiliation(s)
- Mihai Octavian Dan
- Department of Microbiology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Daniela Tǎlǎpan
- Department of Microbiology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Microbiology Laboratory, “Matei Bals” National Institute of Infectious Diseases, Bucharest, Romania
| |
Collapse
|
37
|
Nasrollahian S, Graham JP, Halaji M. A review of the mechanisms that confer antibiotic resistance in pathotypes of E. coli. Front Cell Infect Microbiol 2024; 14:1387497. [PMID: 38638826 PMCID: PMC11024256 DOI: 10.3389/fcimb.2024.1387497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024] Open
Abstract
The dissemination of antibiotic resistance in Escherichia coli poses a significant threat to public health worldwide. This review provides a comprehensive update on the diverse mechanisms employed by E. coli in developing resistance to antibiotics. We primarily focus on pathotypes of E. coli (e.g., uropathogenic E. coli) and investigate the genetic determinants and molecular pathways that confer resistance, shedding light on both well-characterized and recently discovered mechanisms. The most prevalent mechanism continues to be the acquisition of resistance genes through horizontal gene transfer, facilitated by mobile genetic elements such as plasmids and transposons. We discuss the role of extended-spectrum β-lactamases (ESBLs) and carbapenemases in conferring resistance to β-lactam antibiotics, which remain vital in clinical practice. The review covers the key resistant mechanisms, including: 1) Efflux pumps and porin mutations that mediate resistance to a broad spectrum of antibiotics, including fluoroquinolones and aminoglycosides; 2) adaptive strategies employed by E. coli, including biofilm formation, persister cell formation, and the activation of stress response systems, to withstand antibiotic pressure; and 3) the role of regulatory systems in coordinating resistance mechanisms, providing insights into potential targets for therapeutic interventions. Understanding the intricate network of antibiotic resistance mechanisms in E. coli is crucial for the development of effective strategies to combat this growing public health crisis. By clarifying these mechanisms, we aim to pave the way for the design of innovative therapeutic approaches and the implementation of prudent antibiotic stewardship practices to preserve the efficacy of current antibiotics and ensure a sustainable future for healthcare.
Collapse
Affiliation(s)
- Sina Nasrollahian
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jay P. Graham
- Environmental Health Sciences Division, School of Public Health, University of California, Berkeley, CA, United States
| | - Mehrdad Halaji
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Microbiology and Biotechnology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
38
|
Li J, Liao Q, Wang Y, Wang X, Liu J, Zha R, He JZ, Zhang M, Zhang W. Involvement of functional metabolism promotes the enrichment of antibiotic resistome in drinking water: Based on the PICRUSt2 functional prediction. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 356:120544. [PMID: 38471323 DOI: 10.1016/j.jenvman.2024.120544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
Biofilters are the important source and sink of antibiotic resistance genes (ARGs) and antibiotic resistance bacteria (ARB) in the drinking water. Current studies generally ascribed the prevalence of BAR in biofilter from the perspective of gene behavior, i.e. horizontal gene transfer (HGT), little attentions have been paid on the ARGs carrier- ARB. In this study, we proposed the hypothesis that ARB participating in pollutant metabolism processes and becoming dominant is an important way for the enrichment of ARGs. To verify this, the antibiotic resistome and bacterial functional metabolic pathways of a sand filter was profiled using heterotrophic bacterial plate counting method (HPC), high-throughput qPCR, Illumina Hiseq sequencing and PICRUSt2 functional prediction. The results illustrated a significant leakage of ARB in the effluent of the sand filter with an average absolute abundance of approximately 102-103 CFU/mL. Further contribution analysis revealed that the dominant genera, such as Acinetobacter spp., Aeromonas spp., Elizabethkingia spp., and Bacillus spp., were primary ARGs hosts, conferring resistance to multiple antibiotics including sulfamethoxazole, tetracycline and β-lactams. Notably, these ARGs hosts were involved in nitrogen metabolism, including extracellular nitrate/nitrite transport and nitrite reduction, which are crucial in nitrification and denitrification in biofilters. For example, Acinetobacter spp., the dominant bacteria in the filter (relative abundance 69.97 %), contributed the majority of ARGs and 53.79 % of nitrite reduction function. That is, ARB can predominate by participating in the nitrogen metabolism pathways, facilitating the enrichment of ARGs. These findings provide insights into the stable presence of ARGs in biofilters from a functional metabolism perspective, offering a significant supplementary to the mechanisms of the emergence, maintenance, and transmission of BARin drinking water.
Collapse
Affiliation(s)
- Jiabing Li
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Qiuyu Liao
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Yun Wang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Xuansen Wang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Jinchi Liu
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| | - Ruibo Zha
- School of Cultural Tourism and Public Administration, Fujian Normal University, Fuzhou 350117, China
| | - Ji-Zheng He
- Key Laboratory for Humid Subtropical Eco-geographical Processes of the Ministry of Education, Fujian Normal University, Fuzhou 350007, China; Fujian Sanming Forest Ecosystem National Observation and Research Station, Fujian Normal University, Sanming 365002, China
| | - Menglu Zhang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Key Laboratory for Humid Subtropical Eco-geographical Processes of the Ministry of Education, Fujian Normal University, Fuzhou 350007, China; Fujian Sanming Forest Ecosystem National Observation and Research Station, Fujian Normal University, Sanming 365002, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China.
| | - Weifang Zhang
- College of Environmental and Resource Science, Fujian Normal University, Fuzhou 350117, China; Fujian Provincial Key Laboratory of Pollution Control & Resource Reuse (Fujian Normal University) Fuzhou 350117, China
| |
Collapse
|
39
|
Alvernaz SA, Wenzel ES, Nagelli U, Pezley LB, LaBomascus B, Gilbert JA, Maki PM, Tussing-Humphreys L, Peñalver Bernabé B. Inflammatory Dietary Potential Is Associated with Vitamin Depletion and Gut Microbial Dysbiosis in Early Pregnancy. Nutrients 2024; 16:935. [PMID: 38612969 PMCID: PMC11013194 DOI: 10.3390/nu16070935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
Pregnancy alters many physiological systems, including the maternal gut microbiota. Diet is a key regulator of this system and can alter the host immune system to promote inflammation. Multiple perinatal disorders have been associated with inflammation, maternal metabolic alterations, and gut microbial dysbiosis, including gestational diabetes mellitus, pre-eclampsia, preterm birth, and mood disorders. However, the effects of high-inflammatory diets on the gut microbiota during pregnancy have yet to be fully explored. We aimed to address this gap using a system-based approach to characterize associations among dietary inflammatory potential, a measure of diet quality, and the gut microbiome during pregnancy. Forty-seven pregnant persons were recruited prior to 16 weeks of gestation. Participants completed a food frequency questionnaire (FFQ) and provided fecal samples. Dietary inflammatory potential was assessed using the Dietary Inflammatory Index (DII) from the FFQ data. Fecal samples were analyzed using 16S rRNA amplicon sequencing. Differential taxon abundances with respect to the DII score were identified, and the microbial metabolic potential was predicted using PICRUSt2. Inflammatory diets were associated with decreased vitamin and mineral intake and a dysbiotic gut microbiota structure and predicted metabolism. Gut microbial compositional differences revealed a decrease in short-chain fatty acid producers such as Faecalibacterium, and an increase in predicted vitamin B12 synthesis, methylglyoxal detoxification, galactose metabolism, and multidrug efflux systems in pregnant individuals with increased DII scores. Dietary inflammatory potential was associated with a reduction in the consumption of vitamins and minerals and predicted gut microbiota metabolic dysregulation.
Collapse
Affiliation(s)
- Suzanne A. Alvernaz
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
| | - Elizabeth S. Wenzel
- Department of Psychology, University of Illinois, Chicago, IL 60607, USA; (E.S.W.); (P.M.M.)
| | - Unnathi Nagelli
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
| | - Lacey B. Pezley
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Bazil LaBomascus
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Jack A. Gilbert
- Department of Pediatrics, University of California, San Diego, CA 92093, USA;
- Scripps Oceanographic Institute, University of California, San Diego, CA 92037, USA
| | - Pauline M. Maki
- Department of Psychology, University of Illinois, Chicago, IL 60607, USA; (E.S.W.); (P.M.M.)
- Department of Psychiatry, University of Illinois, Chicago, IL 60612, USA
- Department of Obstetrics and Gynecology, University of Illinois, Chicago, IL 60612, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
- Center for Bioinformatics and Quantitative Biology, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
40
|
Sahoo M, Behera DU, Sahoo RK, Sahoo S, Dey S, Subudhi E. Synergistic action of 6-gingerol as an adjuvant to colistin for susceptibility enhancement in multidrug-resistant Klebsiella pneumoniae isolates. RSC Adv 2024; 14:7779-7785. [PMID: 38444972 PMCID: PMC10912974 DOI: 10.1039/d3ra07835c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/18/2024] [Indexed: 03/07/2024] Open
Abstract
The growing threat to human health posed by multidrug-resistant Klebsiella pneumoniae (MDR-KP) indicates an urgent need to develop alternative therapeutic options. The emergence of colistin resistance further adds to the complexity. The study aims to explore in silico-screened phytomolecule 6-gingerol, the most potent active constituent of ginger, as an adjuvant to restore sensitivity in MDR-KP isolates to colistin. The screening of phytocompounds of Zingiber officinale were obtained from the spiceRx database, and molecular docking with efflux pump protein AcrB was performed using Schrödinger's Glide program. The synergistic and bactericidal effects of 6-gingerol in combination with colistin against MDR-KP isolates were determined following broth micro-dilution (MIC), checkerboard assay, and time-kill study. 6-Gingerol showed a good binding affinity with AcrB protein (-9.32 kcal mol-1) and followed the Lipinski rule of (RO5), demonstrating favourable drug-like properties. Further, the synergistic interaction of 6-gingerol with colistin observed from checkerboard assays against efflux-mediated colistin resistance MDR-KP isolates reveals it to be a prospectus adjuvant. The time-killing assays showed the effect of 6-gingerol in combination with colistin to be bactericidal against MSK9 and bacteriostatic against MSK4 and MSK7. Overall, the study provides insights into the potential use of 6-gingerol as a safe and easily available natural product to treat multidrug-resistant K. pneumoniae infections combined with colistin but needs in vivo toxicity evaluation before further recommendations can be made.
Collapse
Affiliation(s)
- Maheswata Sahoo
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| | - Dibyajyoti Uttameswar Behera
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| | - Rajesh Kumar Sahoo
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| | - Saubhagini Sahoo
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| | - Suchanda Dey
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| | - Enketeswara Subudhi
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinga Nagar, Ghatikia Bhubaneswar 751003 Odisha India +91-9861075829
| |
Collapse
|
41
|
Chen Y, Jiang Y, Xue T, Cheng J. Strategies for the eradication of intracellular bacterial pathogens. Biomater Sci 2024; 12:1115-1130. [PMID: 38284808 DOI: 10.1039/d3bm01498c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Intracellular pathogens affect a significant portion of world population and cause millions of deaths each year. They can invade host cells and survive inside them and are extremely resistant to immune systems and antibiotics. Current treatments have limitations, and therefore, new effective therapies are needed to combat this ongoing health challenge. Active research efforts have been made to develop many new strategies to eradicate these intracellular pathogens. In this review, we focus on the intracellular bacterial pathogens and first introduce several representative intracellular bacteria and the diseases they cause. We then discuss the challenges in eradicating these bacteria and summarize the current therapeutics for intracellular bacteria. Finally, recent advances in intracellular bacteria eradication are highlighted.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yunjiang Jiang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518071, China
| | - Tianrui Xue
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Biomaterials and Drug Delivery Laboratory, School of Engineering, Westlake University, Hangzhou 310024, China
| |
Collapse
|
42
|
Liu Y, Van Horn AM, Pham MTN, Dinh BNN, Chen R, Raphael SDR, Paulino A, Thaker K, Somadder A, Frost DJ, Menke CC, Slimak ZC, Slonczewski JL. Fitness trade-offs of multidrug efflux pumps in Escherichia coli K-12 in acid or base, and with aromatic phytochemicals. Appl Environ Microbiol 2024; 90:e0209623. [PMID: 38289137 PMCID: PMC10880634 DOI: 10.1128/aem.02096-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Multidrug efflux pumps are the frontline defense mechanisms of Gram-negative bacteria, yet little is known of their relative fitness trade-offs under gut conditions such as low pH and the presence of antimicrobial food molecules. Low pH contributes to the proton-motive force (PMF) that drives most efflux pumps. We show how the PMF-dependent pumps AcrAB-TolC, MdtEF-TolC, and EmrAB-TolC undergo selection at low pH and in the presence of membrane-permeant phytochemicals. Competition assays were performed by flow cytometry of co-cultured Escherichia coli K-12 strains possessing or lacking a given pump complex. All three pumps showed negative selection under conditions that deplete PMF (pH 5.5 with carbonyl cyanide 3-chlorophenylhydrazone or at pH 8.0). At pH 5.5, selection against AcrAB-TolC was increased by aromatic acids, alcohols, and related phytochemicals such as methyl salicylate. The degree of fitness cost for AcrA was correlated with the phytochemical's lipophilicity (logP). Methyl salicylate and salicylamide selected strongly against AcrA, without genetic induction of drug resistance regulons. MdtEF-TolC and EmrAB-TolC each had a fitness cost at pH 5.5, but salicylate or benzoate made the fitness contribution positive. Pump fitness effects were not explained by gene expression (measured by digital PCR). Between pH 5.5 and 8.0, acrA and emrA were upregulated in the log phase, whereas mdtE expression was upregulated in the transition-to-stationary phase and at pH 5.5 in the log phase. Methyl salicylate did not affect pump gene expression. Our results suggest that lipophilic non-acidic molecules select against a major efflux pump without inducing antibiotic resistance regulons.IMPORTANCEFor drugs that are administered orally, we need to understand how ingested phytochemicals modulate drug resistance in our gut microbiome. Bacteria maintain low-level resistance by proton-motive force (PMF)-driven pumps that efflux many different antibiotics and cell waste products. These pumps play a key role in bacterial defense by conferring resistance to antimicrobial agents at first exposure while providing time for a pathogen to evolve resistance to higher levels of the antibiotic exposed. Nevertheless, efflux pumps confer energetic costs due to gene expression and pump energy expense. The bacterial PMF includes the transmembrane pH difference (ΔpH), which may be depleted by permeant acids and membrane disruptors. Understanding the fitness costs of efflux pumps may enable us to develop resistance breakers, that is, molecules that work together with antibiotics to potentiate their effect. Non-acidic aromatic molecules have the advantage that they avoid the Mar-dependent induction of regulons conferring other forms of drug resistance. We show that different pumps have distinct selection criteria, and we identified non-acidic aromatic molecules as promising candidates for drug resistance breakers.
Collapse
Affiliation(s)
- Yangyang Liu
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | | | - Rachel Chen
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | - Kavya Thaker
- Department of Biology, Kenyon College, Gambier, Ohio, USA
| | | | | | | | | | | |
Collapse
|
43
|
Padhi AK, Maurya S. Uncovering the secrets of resistance: An introduction to computational methods in infectious disease research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:173-220. [PMID: 38448135 DOI: 10.1016/bs.apcsb.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Antimicrobial resistance (AMR) is a growing global concern with significant implications for infectious disease control and therapeutics development. This chapter presents a comprehensive overview of computational methods in the study of AMR. We explore the prevalence and statistics of AMR, underscoring its alarming impact on public health. The role of AMR in infectious disease outbreaks and its impact on therapeutics development are discussed, emphasizing the need for novel strategies. Resistance mutations are pivotal in AMR, enabling pathogens to evade antimicrobial treatments. We delve into their importance and contribution to the spread of AMR. Experimental methods for quantitatively evaluating resistance mutations are described, along with their limitations. To address these challenges, computational methods provide promising solutions. We highlight the advantages of computational approaches, including rapid analysis of large datasets and prediction of resistance profiles. A comprehensive overview of computational methods for studying AMR is presented, encompassing genomics, proteomics, structural bioinformatics, network analysis, and machine learning algorithms. The strengths and limitations of each method are briefly outlined. Additionally, we introduce ResScan-design, our own computational method, which employs a protein (re)design protocol to identify potential resistance mutations and adaptation signatures in pathogens. Case studies are discussed to showcase the application of ResScan in elucidating hotspot residues, understanding underlying mechanisms, and guiding the design of effective therapies. In conclusion, we emphasize the value of computational methods in understanding and combating AMR. Integration of experimental and computational approaches can expedite the discovery of innovative antimicrobial treatments and mitigate the threat posed by AMR.
Collapse
Affiliation(s)
- Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India.
| | - Shweata Maurya
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
44
|
Sahoo P. Complementary supramolecular drug associates in perfecting the multidrug therapy against multidrug resistant bacteria. Front Immunol 2024; 15:1352483. [PMID: 38415251 PMCID: PMC10897028 DOI: 10.3389/fimmu.2024.1352483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
The inappropriate and inconsistent use of antibiotics in combating multidrug-resistant bacteria exacerbates their drug resistance through a few distinct pathways. Firstly, these bacteria can accumulate multiple genes, each conferring resistance to a specific drug, within a single cell. This accumulation usually takes place on resistance plasmids (R). Secondly, multidrug resistance can arise from the heightened expression of genes encoding multidrug efflux pumps, which expel a broad spectrum of drugs from the bacterial cells. Additionally, bacteria can also eliminate or destroy antibiotic molecules by modifying enzymes or cell walls and removing porins. A significant limitation of traditional multidrug therapy lies in its inability to guarantee the simultaneous delivery of various drug molecules to a specific bacterial cell, thereby fostering incremental drug resistance in either of these paths. Consequently, this approach prolongs the treatment duration. Rather than using a biologically unimportant coformer in forming cocrystals, another drug molecule can be selected either for protecting another drug molecule or, can be selected for its complementary activities to kill a bacteria cell synergistically. The development of a multidrug cocrystal not only improves tabletability and plasticity but also enables the simultaneous delivery of multiple drugs to a specific bacterial cell, philosophically perfecting multidrug therapy. By adhering to the fundamental tenets of multidrug therapy, the synergistic effects of these drug molecules can effectively eradicate bacteria, even before they have the chance to develop resistance. This approach has the potential to shorten treatment periods, reduce costs, and mitigate drug resistance. Herein, four hypotheses are presented to create complementary drug cocrystals capable of simultaneously reaching bacterial cells, effectively destroying them before multidrug resistance can develop. The ongoing surge in the development of novel drugs provides another opportunity in the fight against bacteria that are constantly gaining resistance to existing treatments. This endeavour holds the potential to combat a wide array of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Pathik Sahoo
- International Center for Materials and Nanoarchitectronics (MANA), Research Center for Advanced Measurement and Characterization (RCAMC), National Institute for Materials Science, Tsukuba, Japan
- Foundation of Physics Research Center (FoPRC), Celico, Italy
| |
Collapse
|
45
|
Wang Y, Wu Z, Wang Z, Du H, Xiao S, Lu L, Wang Z. Analyses of the Antibiofilm Activity of o-Phenanthroline Monohydrate against Enterococcus faecalis and Staphylococcus aureus and the Mechanisms Underlying These Effects. ACS Infect Dis 2024; 10:638-649. [PMID: 38258383 DOI: 10.1021/acsinfecdis.3c00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Enterococcus faecalis and Staphylococcus aureus exhibit robust biofilm formation capabilities, the formation of which is closely linked to pathogenicity and drug resistance, thereby resulting in host infection and treatment failure. o-Phenanthroline monohydrate (o-Phen) and its derivatives demonstrate a wide range of antibacterial and antifungal activities. In this study, we aimed to explore the antibiofilm activity of o-Phen to E. faecalis and S. aureus and provide insights into the molecular mechanisms for combating biofilm resistance. We demonstrated that o-Phen possesses significant antibacterial and antibiofilm properties against E. faecalis and S. aureus, inducing alterations in bacterial morphology, compromising cell membrane integrity, and exhibiting synergistic effects with β-lactam antibiotics at sub-MIC concentrations. The adhesion ability and automatic condensation capacity of, and synthesis of, extracellular polymers by E. faecalis cells were reduced by o-Phen, resulting in the inhibition of biofilm formation. Importantly, transcriptome analysis revealed 354 upregulated and 456 downregulated genes in o-Phen-treated E. faecalis. Differentially expressed genes were enriched in 11 metabolism-related pathways, including amino acid metabolism, pyrimidine metabolism, and glycolysis/gluconeogenesis. Moreover, the oppA, CeuA, and ZnuB genes involved in the ABC transport system, and the PBP1A penicillin-binding protein-coding genes sarA and mrcA were significantly downregulated. The multidrug efflux pump system and membrane permeability genes mdtG and hlyD, and bacterial adhesion-related genes, including adcA and fss2 were also downregulated, while mraZ and ASP23 were upregulated. Thus, o-Phen is anticipated to be an effective alternative drug for the treatment of E. faecalis and S. aureus biofilm-associated infections.
Collapse
Affiliation(s)
- Yu Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Zhouhui Wu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Zhiwen Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Heng Du
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Shuang Xiao
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Lin Lu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Zhen Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| |
Collapse
|
46
|
Grooters KE, Ku JC, Richter DM, Krinock MJ, Minor A, Li P, Kim A, Sawyer R, Li Y. Strategies for combating antibiotic resistance in bacterial biofilms. Front Cell Infect Microbiol 2024; 14:1352273. [PMID: 38322672 PMCID: PMC10846525 DOI: 10.3389/fcimb.2024.1352273] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Biofilms, which are complexes of microorganisms that adhere to surfaces and secrete protective extracellular matrices, wield substantial influence across diverse domains such as medicine, industry, and environmental science. Despite ongoing challenges posed by biofilms in clinical medicine, research in this field remains dynamic and indeterminate. This article provides a contemporary assessment of biofilms and their treatment, with a focus on recent advances, to chronicle the evolving landscape of biofilm research.
Collapse
Affiliation(s)
- Kayla E. Grooters
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Jennifer C. Ku
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - David M. Richter
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Matthew J. Krinock
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Ashley Minor
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Patrick Li
- University of Michigan, Ann Arbor, MI, United States
- Division of Biomedical Engineering, Department of Orthopedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Audrey Kim
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Robert Sawyer
- Department of Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Yong Li
- Division of Biomedical Engineering, Department of Orthopedic Surgery, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
47
|
Alvernaz SA, Wenzel ES, Nagelli U, Pezley LB, LaBomascus B, Gilbert JA, Maki PM, Tussing-Humphreys L, Peñalver Bernabé B. Inflammatory dietary potential is associated with vitamin depletion and gut microbial dysbiosis in early pregnancy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.12.02.23299325. [PMID: 38076865 PMCID: PMC10705629 DOI: 10.1101/2023.12.02.23299325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Background Pregnancy alters many physiological systems, including the maternal gut microbiota. Diet is a key regulator of this system and can alter the host immune system to promote inflammation. Multiple perinatal disorders have been associated with inflammation, maternal metabolic alterations, and gut microbial dysbiosis, including gestational diabetes mellitus, preeclampsia, preterm birth, and mood disorders. However, the effects of high inflammatory diets on the gut microbiota during pregnancy have yet to be fully explored. Objective To use a systems-based approach to characterize associations among dietary inflammatory potential, a measure of diet quality, and the gut microbiome during pregnancy. Methods Forty-nine pregnant persons were recruited prior to 16 weeks of gestation. Participants completed a food frequency questionnaire (FFQ) and provided fecal samples. Dietary inflammatory potential was assessed using the Dietary Inflammatory Index (DII) from FFQ data. Fecal samples were analyzed using 16S rRNA amplicon sequencing. Differential taxon abundance with respect to DII score were identified, and microbial metabolic potential was predicted using PICRUSt2. Results Inflammatory diets were associated with decreased vitamin and mineral intake and dysbiotic gut microbiota structure and predicted metabolism. Gut microbial compositional differences revealed a decrease in short chain fatty acid producers such as Faecalibacterium, and an increase in predicted vitamin B12 synthesis, methylglyoxal detoxification, galactose metabolism and multi drug efflux systems in pregnant individuals with increased DII scores. Conclusions Dietary inflammatory potential was associated with a reduction in the consumption of vitamins & minerals and predicted gut microbiota metabolic dysregulation.
Collapse
Affiliation(s)
- Suzanne A. Alvernaz
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | | | - Unnathi Nagelli
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | - Lacey B. Pezley
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL, USA
| | - Bazil LaBomascus
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL, USA
| | - Jack A. Gilbert
- Department of Pediatrics, University of California, San Diego, CA, USA
- Scripps Oceanographic Institute, University of California, San Diego, CA, USA
| | - Pauline M. Maki
- Department of Psychology, University of Illinois, Chicago, IL, USA
- Department of Psychiatry, University of Illinois, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois, Chicago, IL, USA
| | | | - Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois, Chicago, IL, USA
| |
Collapse
|
48
|
Kaur M, Arya P, Chosyang S, Singh B. Comprehending conformational changes in EmrE, multidrug transporter at different pH: insights from molecular dynamics simulations. J Biomol Struct Dyn 2024:1-14. [PMID: 38180013 DOI: 10.1080/07391102.2023.2298386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
EmrE is a small multidrug resistance (SMR) pump of antiparallel topology that confers resistance to a broad range of polyaromatic cations in Escherichia coli. Atomic-level understanding of conformational changes for the selectivity of substrate and transport of a diverse array of drugs through the smallest known efflux pumps is crucial to multi-drug resistance. Therefore, the present study aims to provide insights into conformational changes during the transport through EmrE transporter at different pH. Molecular dynamics simulations have been carried out on the complete structure of EmrE in the absence of substrate. Computational analyses such as secondary structure, principal component, dynamic cross-correlation matrix, and hydrogen bond calculations have been performed. Analysis of MD trajectories in this study revealed pH-dependent interactions that influenced the structural dynamics of EmrE. Notably, at high pH, Glu14 and Tyr60 in both monomers formed electrostatic interactions, while these interactions decreased significantly at a low pH. Interestingly, a kink at helix 3 (H3) and dual open conformation of EmrE at low pH were also observed in contrast to a closed state discerned towards the periplasmic side at high pH. Significant interactions between C-terminal residues and residues at the edge of H1 & Loop1 and H3 & Loop3 were identified, suggesting their role in stabilizing the closed conformation of EmrE at the periplasmic end under high pH conditions. The present study enhances our understanding of EmrE's conformational changes, shedding light on the pH-dependent mechanisms that are likely to impact its function in multi-drug resistance.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manpreet Kaur
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Preeti Arya
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- Faculty of Allied Health Sciences, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Stanzin Chosyang
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Balvinder Singh
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
49
|
Jacob TV, Doshi GM. A Mini-review on Helicobacter pylori with Gastric Cancer and Available Treatments. Endocr Metab Immune Disord Drug Targets 2024; 24:277-290. [PMID: 37622707 DOI: 10.2174/1871530323666230824161901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Helicobacter pylori (H. pylori) is the most thoroughly researched etiological component for stomach inflammation and malignancies. Even though there are conventional recommendations and treatment regimens for eradicating H. pylori, failure rates continue to climb. Antibiotic resistance contributes significantly to misdiagnoses, false positive results, and clinical failures, all of which raise the chance of infection recurrence. This review aims to explore the molecular mechanisms underlying drug resistance in H. pylori and discuss novel approaches for detecting genotypic resistance. Modulation of drug uptake/ efflux, biofilm, and coccoid development. Newer genome sequencing approaches capable of detecting H. pylori genotypic resistance are presented. Prolonged infection in the stomach causes major problems such as gastric cancer. The review discusses how H. pylori causes stomach cancer, recent biomarkers such as miRNAs, molecular pathways in the development of gastric cancer, and diagnostic methods and clinical trials for the disease. Efforts have been made to summarize the recent advancements made toward early diagnosis and novel therapeutic approaches for H. pylori-induced gastric cancer.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
50
|
Druzhkova I, Komarova A, Nikonova E, Baigildin V, Mozherov A, Shakirova Y, Lisitsa U, Shcheslavskiy V, Ignatova N, Shirshin E, Shirmanova M, Tunik S. Monitoring the Intracellular pH and Metabolic State of Cancer Cells in Response to Chemotherapy Using a Combination of Phosphorescence Lifetime Imaging Microscopy and Fluorescence Lifetime Imaging Microscopy. Int J Mol Sci 2023; 25:49. [PMID: 38203221 PMCID: PMC10779161 DOI: 10.3390/ijms25010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
The extracellular matrix (ECM), in which collagen is the most abundant protein, impacts many aspects of tumor physiology, including cellular metabolism and intracellular pH (pHi), as well as the efficacy of chemotherapy. Meanwhile, the role of collagen in differential cell responses to treatment within heterogeneous tumor environments remains poorly investigated. In the present study, we simultaneously monitored the changes in pHi and metabolism in living colorectal cancer cells in vitro upon treatment with a chemotherapeutic combination, FOLFOX (5-fluorouracil, oxaliplatin and leucovorin). The pHi was followed using the new pH-sensitive probe BC-Ga-Ir, working in the mode of phosphorescence lifetime imaging (PLIM), and metabolism was assessed from the autofluorescence of the metabolic cofactor NAD(P)H using fluorescence lifetime imaging (FLIM) with a two-photon laser scanning microscope. To model the ECM, 3D collagen-based hydrogels were used, and comparisons with conventional monolayer cells were made. It was found that FOLFOX treatment caused an early temporal intracellular acidification (reduction in pHi), followed by a shift to more alkaline values, and changed cellular metabolism to a more oxidative state. The presence of unstructured collagen markedly reduced the cytotoxic effects of FOLFOX, and delayed and diminished the pHi and metabolic responses. These results support the observation that collagen is a factor in the heterogeneous response of cancer cells to chemotherapy and a powerful regulator of their metabolic behavior.
Collapse
Affiliation(s)
- Irina Druzhkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Anastasiya Komarova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia
| | - Elena Nikonova
- Laboratory of Clinical Biophotonics, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (E.N.); (E.S.)
| | - Vadim Baigildin
- Institute of Chemistry, Saint-Petersburg State University, 198504 St. Petersburg, Russia; (V.B.); (Y.S.)
| | - Artem Mozherov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Yuliya Shakirova
- Institute of Chemistry, Saint-Petersburg State University, 198504 St. Petersburg, Russia; (V.B.); (Y.S.)
| | - Uliana Lisitsa
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Vladislav Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Nadezhda Ignatova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Evgeny Shirshin
- Laboratory of Clinical Biophotonics, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (E.N.); (E.S.)
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marina Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia; (A.K.); (A.M.); (U.L.); (V.S.); (N.I.); (M.S.)
| | - Sergey Tunik
- Institute of Chemistry, Saint-Petersburg State University, 198504 St. Petersburg, Russia; (V.B.); (Y.S.)
| |
Collapse
|