Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (38)

Search Parameters:
Keywords = CRISPR-based antimicrobials

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
42 pages, 1647 KB  
Review
Preventive Immunology for Livestock and Zoonotic Infectious Diseases in the One Health Era: From Mechanistic Insights to Innovative Interventions
by Eman Marzouk and Ahmed I. Alajaji
Vet. Sci. 2025, 12(10), 1014; https://doi.org/10.3390/vetsci12101014 - 20 Oct 2025
Viewed by 638
Abstract
Preventive immunology is emerging as a cornerstone of animal infectious disease control within One Health, shifting emphasis from treatment to prevention. This review integrates mechanistic insights in host immunity with a comparative evaluation of next-generation interventions—mRNA/DNA and viral-vector vaccines, nanovaccines, monoclonal antibodies, cytokine [...] Read more.
Preventive immunology is emerging as a cornerstone of animal infectious disease control within One Health, shifting emphasis from treatment to prevention. This review integrates mechanistic insights in host immunity with a comparative evaluation of next-generation interventions—mRNA/DNA and viral-vector vaccines, nanovaccines, monoclonal antibodies, cytokine modulators, probiotics/postbiotics, bacteriophages, and CRISPR-based approaches—highlighting their immunogenicity, thermostability, delivery, and field readiness. Distinct from prior reviews, we appraise diagnostics as preventive tools (point-of-care assays, biosensors, MALDI-TOF MS, AI-enabled analytics) that enable early detection, risk prediction, and targeted interventions, and we map quantifiable links between successful prevention and reduced antimicrobial use. We embed translation factors—regulatory alignment, scalable manufacturing, workforce capacity, equitable access in LMICs, and public trust—alongside environmental and zoonotic interfaces that shape antimicrobial resistance dynamics. We also provide a critical analysis of limitations and failure cases: gene editing may require stacked edits and concurrent vaccination; phage programs must manage host range, resistance, stability, and regulation; and probiotic benefits remain context-specific. Finally, we present a risk–benefit–readiness framework and a time-bound research agenda to guide deployment and evaluation across animal–human–environmental systems. Coordinating scientific innovation with governance and ethics can measurably reduce disease burden, curb antimicrobial consumption, and improve health outcomes across species. Full article
Show Figures

Figure 1

15 pages, 325 KB  
Review
Polymicrobial Infections: A Comprehensive Review on Current Context, Diagnostic Bottlenecks and Future Directions
by Amit Patnaik, Titirsha Kayal and Soumya Basu
Acta Microbiol. Hell. 2025, 70(4), 39; https://doi.org/10.3390/amh70040039 - 14 Oct 2025
Viewed by 406
Abstract
Worldwide, polymicrobial infections (PMIs) account for an estimated 20–50% of severe clinical infection cases, with biofilm-associated and device-related infections reaching 60–80% in hospitalized patients. This review discusses the clinical burden of major infections in which PMIs are almost inevitable, such as diabetic foot [...] Read more.
Worldwide, polymicrobial infections (PMIs) account for an estimated 20–50% of severe clinical infection cases, with biofilm-associated and device-related infections reaching 60–80% in hospitalized patients. This review discusses the clinical burden of major infections in which PMIs are almost inevitable, such as diabetic foot infections, intra-abdominal infections, pneumonia, and biofilm-associated device infections. Globally, the PMI landscape is diverse; however, the Indian subcontinent is a PMI hotspot where high comorbidities, endemic antimicrobial resistance, and underdeveloped diagnostic capacity elevate the risks of poor outcomes. Existing diagnostic like culture-based methods, PCR panels, sequencing, and biomarker-based assays are constrained by sensitivity, turnaround times (TATs), and high costs. Vulnerable populations, particularly neonates, the elderly, immunocompromised patients, and socioeconomically marginalized groups, show case-fatality rates 2-fold higher than monomicrobial infections in similar settings. Emerging diagnostic solutions include CRISPR-based multiplex assays, artificial intelligence-based metagenomic platforms, and sensitive biosensors with point-of-care applicability. These technologies show potential in reducing the TAT (<2 h) with high accuracy (>95%). However, their translation to real-world settings depends critically on affordability, integration into healthcare pathways, and supportive policy. This will provide equitable diagnostic access, particularly in low- and middle-income countries (LMICs). Full article
24 pages, 1623 KB  
Review
Beyond the Resistome: Molecular Insights, Emerging Therapies, and Environmental Drivers of Antibiotic Resistance
by Nada M. Nass and Kawther A. Zaher
Antibiotics 2025, 14(10), 995; https://doi.org/10.3390/antibiotics14100995 - 4 Oct 2025
Viewed by 493
Abstract
Antibiotic resistance remains one of the most formidable challenges to modern medicine, threatening to outpace therapeutic innovation and undermine decades of clinical progress. While resistance was once viewed narrowly as a clinical phenomenon, it is now understood as the outcome of complex ecological [...] Read more.
Antibiotic resistance remains one of the most formidable challenges to modern medicine, threatening to outpace therapeutic innovation and undermine decades of clinical progress. While resistance was once viewed narrowly as a clinical phenomenon, it is now understood as the outcome of complex ecological and molecular interactions that span soil, water, agriculture, animals, and humans. Environmental reservoirs act as silent incubators of resistance genes, with horizontal gene transfer and stress-induced mutagenesis fueling their evolution and dissemination. At the molecular level, advances in genomics, structural biology, and systems microbiology have revealed intricate networks involving plasmid-mediated resistance, efflux pump regulation, integron dynamics, and CRISPR-Cas interactions, providing new insights into the adaptability of pathogens. Simultaneously, the environmental dimensions of resistance, from wastewater treatment plants and aquaculture to airborne dissemination, highlight the urgency of adopting a One Health framework. Yet, alongside this growing threat, novel therapeutic avenues are emerging. Innovative β-lactamase inhibitors, bacteriophage-based therapies, engineered lysins, antimicrobial peptides, and CRISPR-driven antimicrobials are redefining what constitutes an “antibiotic” in the twenty-first century. Furthermore, artificial intelligence and machine learning now accelerate drug discovery and resistance prediction, raising the possibility of precision-guided antimicrobial stewardship. This review synthesizes molecular insights, environmental drivers, and therapeutic innovations to present a comprehensive landscape of antibiotic resistance. By bridging ecological microbiology, molecular biology, and translational medicine, it outlines a roadmap for surveillance, prevention, and drug development while emphasizing the need for integrative policies to safeguard global health. Full article
(This article belongs to the Special Issue Antimicrobial Resistance and Environmental Health, 2nd Edition)
Show Figures

Figure 1

23 pages, 1444 KB  
Review
Streptococcus thermophilus: Metabolic Properties, Functional Features, and Useful Applications
by Alyaa Zaidan Ghailan and Alaa Kareem Niamah
Appl. Microbiol. 2025, 5(4), 101; https://doi.org/10.3390/applmicrobiol5040101 - 23 Sep 2025
Viewed by 1741
Abstract
Streptococcus thermophilus is a Gram-positive, homofermentative lactic acid bacterium classified within the Firmicutes phylum, recognized for its probiotic properties and significant role in promoting human health. This review consolidates existing understanding of its metabolic pathways, functional metabolites, and diverse applications, highlighting evidence-based insights [...] Read more.
Streptococcus thermophilus is a Gram-positive, homofermentative lactic acid bacterium classified within the Firmicutes phylum, recognized for its probiotic properties and significant role in promoting human health. This review consolidates existing understanding of its metabolic pathways, functional metabolites, and diverse applications, highlighting evidence-based insights to enhance scientific integrity. S. thermophilus predominantly ferments lactose through the Embden-Meyerhof-Parnas pathway, resulting in L(+)-lactic acid as the primary end-product, along with secondary metabolites including acetic acid, formic acid, and pyruvate derivatives. Exopolysaccharides (EPS) are composed of repeating units of glucose, galactose, rhamnose, and N-acetylgalactosamine. They display strain-specific molecular weights ranging from 10 to 2000 kDa and contribute to the viscosity of fermented products, while also providing antioxidant and immunomodulatory benefits. Aromatic compounds such as acetaldehyde and phenylacetic acid are products of amino acid catabolism and carbohydrate metabolism, playing a significant role in the sensory characteristics observed in dairy fermentations. Bacteriocins, such as thermophilins (e.g., Thermophilin 13, 110), exhibit extensive antimicrobial efficacy against pathogens including Listeria monocytogenes and Bacillus cereus. Their activity is modulated by quorum-sensing mechanisms that involve the blp gene cluster, and they possess significant stability under heat and pH variations, making them suitable for biopreservation applications. In food applications, S. thermophilus functions as a Generally Recognized as Safe (GRAS) starter culture in the production of yogurt and cheese, working in conjunction with Lactobacillus delbrueckii subsp. bulgaricus to enhance acidification and improve texture. Specific strains have been identified to mitigate lactose intolerance, antibiotic-related diarrhea, and inflammatory bowel diseases through the modulation of gut microbiota, the production of short-chain fatty acids, and the inhibition of Helicobacter pylori. The genome, characterized by a G + C content of approximately 37 mol%, facilitates advancements in Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas technology and heterologous protein expression, with applications extending to non-dairy fermentations and the development of postbiotics. This review emphasizes the adaptability of S. thermophilus, showcasing the variability among strains and the necessity for thorough preclinical and clinical validation to fully utilize its potential in health, sustainable agriculture, and innovation. It also addresses challenges such as susceptibility to bacteriophages and limitations in proteolytic activity. Full article
Show Figures

Figure 1

20 pages, 1568 KB  
Review
Probiotics and Postbiotics for Green Control of Foodborne Pathogens: Intelligent Detection and Biopreservation Strategies for Safer Foods
by Alice N. Mafe and Dietrich Büsselberg
Foods 2025, 14(18), 3281; https://doi.org/10.3390/foods14183281 - 22 Sep 2025
Cited by 1 | Viewed by 1652
Abstract
The extensive use of chemical preservatives in the food industry has raised concerns over their association with gut microbiota imbalance, allergenic reactions, and potential carcinogenicity. Growing consumer demand for “clean label” products, coupled with regulatory pressures, has accelerated the search for safer and [...] Read more.
The extensive use of chemical preservatives in the food industry has raised concerns over their association with gut microbiota imbalance, allergenic reactions, and potential carcinogenicity. Growing consumer demand for “clean label” products, coupled with regulatory pressures, has accelerated the search for safer and more sustainable alternatives. In this study, it is reported for the first time that the synthesis of AIEE-type Supra-CDs using p-phenylenediamine (p-PA) and thiourea (TU), a breakthrough that provides a new class of nanomaterials with superior optical and antimicrobial properties. More importantly, the study demonstrates a quantitative improvement of spectral overlap through controllable inner filter effect (IFE), establishing a reliable strategy to enhance detection sensitivity and broaden applicability in food safety monitoring. Beyond their intrinsic antimicrobial potential, these Supra-CDs integrate seamlessly with intelligent detection platforms such as biosensors, CRISPR-based assays, and AI-assisted analytics, enabling real-time evaluation of probiotic- and postbiotic-based preservation systems. By combining novel material synthesis with precision monitoring technologies, this work offers a dual innovation: reducing reliance on synthetic additives while providing scalable tools for sustainable food preservation. The findings not only advance the frontier of biopreservation research but also align with global initiatives for consumer health and environmental sustainability. Full article
Show Figures

Figure 1

16 pages, 1194 KB  
Systematic Review
Biotechnological Innovations to Combat Antimicrobial Resistance and Advance Global Health Equity
by Sima Rugarabamu and Gaspary Mwanyika
Bacteria 2025, 4(3), 46; https://doi.org/10.3390/bacteria4030046 - 5 Sep 2025
Cited by 1 | Viewed by 1136
Abstract
Antimicrobial resistance (AMR) is a growing global health emergency that threatens the effectiveness of modern medicine, exacerbating healthcare costs, morbidity, and mortality, particularly in low- and middle-income countries (LMICs). Traditional approaches to antimicrobial development and stewardship have proven inadequate in curbing the rapid [...] Read more.
Antimicrobial resistance (AMR) is a growing global health emergency that threatens the effectiveness of modern medicine, exacerbating healthcare costs, morbidity, and mortality, particularly in low- and middle-income countries (LMICs). Traditional approaches to antimicrobial development and stewardship have proven inadequate in curbing the rapid emergence and spread of resistant pathogens. This review explores cutting-edge biotechnological innovations as sustainable, precision-based solutions to combat AMR and promote global health equity. A comprehensive narrative review was conducted using literature published between 2018 and 2023 from PubMed, ScienceDirect, and Web of Science. Peer-reviewed studies focusing on novel antimicrobial strategies were thematically analyzed, with attention to efficacy, feasibility, and translational readiness. Key innovations identified include nanotechnology-enhanced antimicrobial delivery, bacteriophage therapy, CRISPR-Cas gene editing, immunotherapy, and personalized medicine. These strategies demonstrated substantial in vitro and in vivo efficacy, such as >90% MRSA biofilm reduction via silver nanoparticles and 95% carbapenem susceptibility restoration in E. coli using CRISPR-Cas9. When integrated with machine learning and rapid diagnostics, these approaches enable precision-targeted therapies and data-informed stewardship, offering scalable solutions adaptable to diverse healthcare systems. Antimicrobial resistance demands urgent, equitable innovation. Integrating biotechnologies like CRISPR, phage therapy, and nanomedicine with data-driven tools offers promising solutions. To ensure real-world impact, we recommend establishing regionally tailored translational research platforms and public–private partnerships as the most effective strategy to scale innovations and strengthen AMR response in low-resource settings. Full article
Show Figures

Figure 1

42 pages, 2279 KB  
Review
From Farm to Fork: Antimicrobial-Resistant Bacterial Pathogens in Livestock Production and the Food Chain
by Ayman Elbehiry and Eman Marzouk
Vet. Sci. 2025, 12(9), 862; https://doi.org/10.3390/vetsci12090862 - 4 Sep 2025
Viewed by 1397
Abstract
Antimicrobial resistance (AMR) in livestock production systems has emerged as a major global health concern, threatening not only animal welfare and agricultural productivity but also food safety and public health. The widespread, and often poorly regulated, use of antimicrobials for growth promotion, prophylaxis, [...] Read more.
Antimicrobial resistance (AMR) in livestock production systems has emerged as a major global health concern, threatening not only animal welfare and agricultural productivity but also food safety and public health. The widespread, and often poorly regulated, use of antimicrobials for growth promotion, prophylaxis, and metaphylaxis has accelerated the emergence and dissemination of resistant bacteria and resistance genes. These elements circulate across interconnected animal, environmental, and human ecosystems, driven by mobile genetic elements and amplified through the food production chain. It is estimated that more than two-thirds of medically important antimicrobials are used in animals, and AMR could cause millions of human deaths annually by mid-century if unchecked. In some livestock systems, multidrug-resistant E. coli prevalence already exceeds half of isolates, particularly in poultry and swine in low- and middle-income countries (LMICs). This narrative review provides a comprehensive overview of the molecular epidemiology, ecological drivers, and One Health implications of AMR in food-producing animals. We highlight key zoonotic and foodborne bacterial pathogens—including Escherichia coli, Salmonella enterica, and Staphylococcus aureus—as well as underappreciated reservoirs in commensal microbiota and livestock environments. Diagnostic platforms spanning phenotypic assays, PCR, MALDI-TOF MS, whole-genome sequencing, and CRISPR-based tools are examined for their roles in AMR detection, surveillance, and resistance gene characterization. We also evaluate current antimicrobial stewardship practices, global and regional surveillance initiatives, and policy frameworks, identifying critical implementation gaps, especially in low- and middle-income countries. Emerging sectors such as aquaculture and insect farming are considered for their potential role as future AMR hotspots. Finally, we outline future directions including real-time genomic surveillance, AI-assisted resistance prediction, and integrated One Health data platforms as essential innovations to combat AMR. Mitigating the threat of AMR in animal agriculture will require coordinated scientific, regulatory, and cross-sectoral responses to ensure the long-term efficacy of antimicrobial agents for both human and veterinary medicine. Full article
Show Figures

Figure 1

29 pages, 2561 KB  
Review
Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release
by Chenxi Huang and Lianghui Cheng
Int. J. Mol. Sci. 2025, 26(17), 8119; https://doi.org/10.3390/ijms26178119 - 22 Aug 2025
Viewed by 853
Abstract
Bioactive peptides encrypted in bovine β-casein display diverse physiological functions, including antihypertensive, antioxidative, antimicrobial, and immunomodulatory activities. These peptides are normally released during gastrointestinal digestion or microbial fermentation, especially by proteolytic systems of lactic acid bacteria (LAB). However, peptide yields vary widely among [...] Read more.
Bioactive peptides encrypted in bovine β-casein display diverse physiological functions, including antihypertensive, antioxidative, antimicrobial, and immunomodulatory activities. These peptides are normally released during gastrointestinal digestion or microbial fermentation, especially by proteolytic systems of lactic acid bacteria (LAB). However, peptide yields vary widely among LAB strains, reflecting strain-specific protease repertoires. To overcome these limitations, the scientific goal of this study is to provide a comprehensive synthesis of how synthetic biology, molecular biotechnology, and systems-level approaches can be leveraged to enhance the targeted discovery and production of β-casein-derived bioactive peptides. Genome engineering tools such as clustered regularly interspaced short palindromic repeats associated system (CRISPR/Cas) systems have been applied to modulate gene expression and metabolic flux in LAB, while inducible expression platforms allow on-demand peptide production. Additionally, cell-free systems based on LAB lysates further provide rapid prototyping for high-throughput screening. Finally, multi-omics approaches, including genomics, transcriptomics, proteomics, and metabolomics, further help pinpoint regulatory bottlenecks and facilitate rational strain optimization. This review provides a comprehensive overview of bioactive peptides derived from bovine β-casein and highlights recent progress in LAB-based strategies—both natural and engineered—for their efficient release. These advances pave the way for developing next-generation functional fermented foods enriched with targeted bioactivities. Full article
Show Figures

Graphical abstract

20 pages, 1527 KB  
Review
From Panels to Pathogen Networks: The Expanding Role of Targeted Sequencing in Veterinary Medicine
by Jiali Luo, Wentao Lu, Ruiting Liu, Shukai Zhang, Jie Cao and Chong Ma
Biology 2025, 14(8), 1075; https://doi.org/10.3390/biology14081075 - 18 Aug 2025
Viewed by 906
Abstract
Targeted sequencing, a pivotal branch of next-generation sequencing (NGS), enables the selective enrichment of specific genomic regions and has demonstrated significant advantages in the detection of animal pathogens. This review systematically explores the underlying principles of targeted sequencing, various enrichment strategies—including PCR amplification, [...] Read more.
Targeted sequencing, a pivotal branch of next-generation sequencing (NGS), enables the selective enrichment of specific genomic regions and has demonstrated significant advantages in the detection of animal pathogens. This review systematically explores the underlying principles of targeted sequencing, various enrichment strategies—including PCR amplification, probe hybridization, and CRISPR-Cas systems—and their key applications in veterinary pathogen diagnostics. Due to its high throughput, sensitivity, and cost-effectiveness, targeted sequencing has been successfully applied in the multiplex detection of pathogens in economically significant livestock, such as cattle, as well as in the surveillance of antimicrobial resistance (AMR) genes, pathogen typing, and source tracing. It is particularly effective in identifying mixed infections and low-abundance pathogens. Nonetheless, wide application is restricted by some factors, like incomprehensive reference databases, cost-effectiveness, and limited application in primary-level laboratories. Further development directions are AI-based panel design, multimodal diagnostic platform integration, standard workflow construction, and introduction of a multi-omics method. Such progress focuses on enhancing the targeted sequencing scalability and precision consistent with the “One Health” initiative objective. Full article
Show Figures

Figure 1

31 pages, 3024 KB  
Review
Synthetic and Functional Engineering of Bacteriophages: Approaches for Tailored Bactericidal, Diagnostic, and Delivery Platforms
by Ola Alessa, Yoshifumi Aiba, Mahmoud Arbaah, Yuya Hidaka, Shinya Watanabe, Kazuhiko Miyanaga, Dhammika Leshan Wannigama and Longzhu Cui
Molecules 2025, 30(15), 3132; https://doi.org/10.3390/molecules30153132 - 25 Jul 2025
Cited by 1 | Viewed by 2685
Abstract
Bacteriophages (phages), the most abundant biological entities on Earth, have long served as both model systems and therapeutic tools. Recent advances in synthetic biology and genetic engineering have revolutionized the capacity to tailor phages with enhanced functionality beyond their natural capabilities. This review [...] Read more.
Bacteriophages (phages), the most abundant biological entities on Earth, have long served as both model systems and therapeutic tools. Recent advances in synthetic biology and genetic engineering have revolutionized the capacity to tailor phages with enhanced functionality beyond their natural capabilities. This review outlines the current landscape of synthetic and functional engineering of phages, encompassing both in-vivo and in-vitro strategies. We describe in-vivo approaches such as phage recombineering systems, CRISPR-Cas-assisted editing, and bacterial retron-based methods, as well as synthetic assembly platforms including yeast-based artificial chromosomes, Gibson, Golden Gate, and iPac assemblies. In addition, we explore in-vitro rebooting using TXTL (transcription–translation) systems, which offer a flexible alternative to cell-based rebooting but are less effective for large genomes or structurally complex phages. Special focus is given to the design of customized phages for targeted applications, including host range expansion via receptor-binding protein modifications, delivery of antimicrobial proteins or CRISPR payloads, and the construction of biocontained, non-replicative capsid systems for safe clinical use. Through illustrative examples, we highlight how these technologies enable the transformation of phages into programmable bactericidal agents, precision diagnostic tools, and drug delivery vehicles. Together, these advances establish a powerful foundation for next-generation antimicrobial platforms and synthetic microbiology. Full article
Show Figures

Figure 1

19 pages, 5087 KB  
Review
Biosensors in Microbial Ecology: Revolutionizing Food Safety and Quality
by Gajanan A. Bodkhe, Vishal Kumar, Xingjie Li, Shichun Pei, Long Ma and Myunghee Kim
Microorganisms 2025, 13(7), 1706; https://doi.org/10.3390/microorganisms13071706 - 21 Jul 2025
Cited by 2 | Viewed by 1885
Abstract
Microorganisms play a crucial role in food processes, safety, and quality through their dynamic interactions with other organisms. In recent years, biosensors have become essential tools for monitoring these processes in the dairy, meat, and fresh produce industries. This review highlights how microbial [...] Read more.
Microorganisms play a crucial role in food processes, safety, and quality through their dynamic interactions with other organisms. In recent years, biosensors have become essential tools for monitoring these processes in the dairy, meat, and fresh produce industries. This review highlights how microbial diversity, starter cultures, and interactions, such as competition and quorum sensing, shape food ecosystems. Diverse biosensor platforms, including electrochemical, optical, piezoelectric, thermal, field-effect transistor-based, and lateral flow assays, offer distinct advantages tailored to specific food matrices and microbial targets, enabling rapid and sensitive detection. Biosensors have been developed for detecting pathogens in real-time monitoring of fermentation and tracking spoilage. Control strategies, including bacteriocins, probiotics, and biofilm management, support food safety, while decontamination methods provide an additional layer of protection. The integration of new techniques, such as nanotechnology, CRISPR, and artificial intelligence, into Internet of Things systems is enhancing precision, particularly in addressing regional food safety challenges. However, their adoption is still hindered by complex food matrices, high costs, and the growing challenge of antimicrobial resistance. Looking ahead, intelligent systems and wearable sensors may help overcome these barriers. Although gaps in standardization and accessibility remain, biosensors are well-positioned to revolutionize food microbiology, linking ecological insights to practical solutions and paving the way for safer, high-quality food worldwide. Full article
(This article belongs to the Special Issue Feature Papers in Food Microbiology)
Show Figures

Figure 1

29 pages, 1254 KB  
Review
Microbial Food Safety and Antimicrobial Resistance in Foods: A Dual Threat to Public Health
by Ayman Elbehiry, Eman Marzouk, Adil Abalkhail, Husam M. Edrees, Abousree T. Ellethy, Abdulaziz M. Almuzaini, Mai Ibrahem, Abdulrahman Almujaidel, Feras Alzaben, Abdullah Alqrni and Akram Abu-Okail
Microorganisms 2025, 13(7), 1592; https://doi.org/10.3390/microorganisms13071592 - 6 Jul 2025
Cited by 4 | Viewed by 4715
Abstract
The intersection of microbial food safety and antimicrobial resistance (AMR) represents a mounting global threat with profound implications for public health, food safety, and sustainable development. This review explores the complex pathways through which foodborne pathogens—such as Salmonella spp., Escherichia coli (E. [...] Read more.
The intersection of microbial food safety and antimicrobial resistance (AMR) represents a mounting global threat with profound implications for public health, food safety, and sustainable development. This review explores the complex pathways through which foodborne pathogens—such as Salmonella spp., Escherichia coli (E. coli), Listeria monocytogenes (L. monocytogenes), and Campylobacter spp.—acquire and disseminate resistance within human, animal, and environmental ecosystems. Emphasizing a One Health framework, we examine the drivers of AMR across sectors, including the misuse of antibiotics in agriculture, aquaculture, and clinical settings, and assess the role of environmental reservoirs in sustaining and amplifying resistance genes. We further discuss the evolution of surveillance systems, regulatory policies, and antimicrobial stewardship programs (ASPs) designed to mitigate resistance across the food chain. Innovations in next-generation sequencing, metagenomics, and targeted therapeutics such as bacteriophage therapy, antimicrobial peptides (AMPs), and CRISPR-based interventions offer promising alternatives to conventional antibiotics. However, the translation of these advances into practice remains uneven, particularly in low- and middle-income countries (LMICs) facing significant barriers to diagnostic access, laboratory capacity, and equitable treatment availability. Our analysis underscores the urgent need for integrated, cross-sectoral action—anchored in science, policy, and education—to curb the global spread of AMR. Strengthening surveillance, investing in research, promoting responsible antimicrobial use, and fostering global collaboration are essential to preserving the efficacy of existing treatments and ensuring the microbiological safety of food systems worldwide. Full article
(This article belongs to the Special Issue Microbial Safety and Beneficial Microorganisms in Foods)
Show Figures

Figure 1

27 pages, 1648 KB  
Review
Carbapenem Resistance in Acinetobacter baumannii: Mechanisms, Therapeutics, and Innovations
by Joyce de Souza, Helena Regina Salomé D’Espindula, Isabel de Farias Ribeiro, Geiziane Aparecida Gonçalves, Marcelo Pillonetto and Helisson Faoro
Microorganisms 2025, 13(7), 1501; https://doi.org/10.3390/microorganisms13071501 - 27 Jun 2025
Cited by 1 | Viewed by 4855
Abstract
The global rise of carbapenem-resistant Acinetobacter baumannii (CRAB) strains poses a critical challenge to healthcare systems due to limited therapeutic options and high mortality rates, especially in intensive care settings. This review explores the epidemiological landscape and molecular mechanisms driving carbapenem resistance, including [...] Read more.
The global rise of carbapenem-resistant Acinetobacter baumannii (CRAB) strains poses a critical challenge to healthcare systems due to limited therapeutic options and high mortality rates, especially in intensive care settings. This review explores the epidemiological landscape and molecular mechanisms driving carbapenem resistance, including the production of diverse beta-lactamases (particularly OXA-type enzymes), porin loss, efflux pump overexpression, and mutations in antibiotic targets. Emerging treatment strategies are discussed, such as the use of new beta-lactam–beta-lactamase inhibitor combinations (e.g., sulbactam–durlobactam), siderophore cephalosporins, next-generation polymyxins, as well as novel agents like zosurabalpin and rifabutin (BV100). Alternative approaches—including phage therapy, antimicrobial peptides, CRISPR-based gene editing, and nanoparticle-based delivery systems—are also evaluated for their potential to bypass traditional resistance mechanisms. Furthermore, advances in artificial intelligence and multi-omics integration are highlighted as tools for identifying novel drug targets and predicting resistance profiles. Together, these innovations represent a multifaceted strategy to overcome CRAB infections, yet their successful implementation requires further clinical validation and coordinated surveillance efforts. This analysis highlights the urgent need for continued investment in innovative treatments and effective resistance monitoring to limit the spread of CRAB and protect the effectiveness of last-line antibiotics. Full article
(This article belongs to the Special Issue Combating Antimicrobial Resistance: Innovations and Strategies)
Show Figures

Figure 1

21 pages, 568 KB  
Review
Armed Phages: A New Weapon in the Battle Against Antimicrobial Resistance
by Cleo Anastassopoulou, Deny Tsakri, Antonios-Periklis Panagiotopoulos, Chrysa Saldari, Antonia P. Sagona and Athanasios Tsakris
Viruses 2025, 17(7), 911; https://doi.org/10.3390/v17070911 - 27 Jun 2025
Cited by 3 | Viewed by 2102
Abstract
The increasing prevalence of multidrug-resistant (MDR) bacterial infections necessitates the exploration of alternative antimicrobial strategies, with phage therapy emerging as a viable option. However, the effectiveness of naturally occurring phages can be significantly limited by bacterial defense systems that include adsorption blocking, restriction–modification, [...] Read more.
The increasing prevalence of multidrug-resistant (MDR) bacterial infections necessitates the exploration of alternative antimicrobial strategies, with phage therapy emerging as a viable option. However, the effectiveness of naturally occurring phages can be significantly limited by bacterial defense systems that include adsorption blocking, restriction–modification, CRISPR-Cas immunity, abortive infection, and NAD+ depletion defense systems. This review examines these bacterial defenses and their implications for phage therapy, while highlighting the potential of phages’ bioengineering to overcome these barriers. By leveraging synthetic biology, genetically engineered phages can be tailored to evade bacterial immunity through such modifications as receptor-binding protein engineering, anti-CRISPR gene incorporation, methylation pattern alterations, and enzymatic degradation of bacterial protective barriers. “Armed phages”, enhanced with antimicrobial peptides, CRISPR-based genome-editing tools, or immune-modulating factors, offer a novel therapeutic avenue. Clinical trials of bioengineered phages, currently SNIPR001 and LBP-EC01, showcase their potential to safely and effectively combat MDR infections. SNIPR001 has completed a Phase I clinical trial evaluating safety in healthy volunteers, while LBP-EC01 is in Phase II trials assessing its performance in the treatment of Escherichia coli-induced urinary tract infections in patients with a history of drug-resistant infections. As “armed phages” progress toward clinical application, they hold great promise for precision-targeted antimicrobial therapies and represent a critical innovation in addressing the global antibiotic resistance crisis. Full article
(This article belongs to the Collection Phage Therapy)
Show Figures

Figure 1

26 pages, 5536 KB  
Review
The Breeding, Cultivation, and Potential Applications of Ornamental Orchids with a Focus on Phalaenopsis—A Brief Review
by Chenjing Han, Fei Dong, Yu Qi, Yenan Wang, Jiao Zhu, Binghai Li, Lijuan Zhang, Xiaohui Lv and Jianghui Wang
Plants 2025, 14(11), 1689; https://doi.org/10.3390/plants14111689 - 31 May 2025
Viewed by 3041
Abstract
The Phalaenopsis genus, a horticulturally vital group within the Orchidaceae, dominates global floriculture markets through strategic cultivar innovation, scalable propagation, and data-driven cultivation. This review systematically examines the breeding, propagation, cultivation management, and potential applications of Phalaenopsis while providing insights into future [...] Read more.
The Phalaenopsis genus, a horticulturally vital group within the Orchidaceae, dominates global floriculture markets through strategic cultivar innovation, scalable propagation, and data-driven cultivation. This review systematically examines the breeding, propagation, cultivation management, and potential applications of Phalaenopsis while providing insights into future research directions. The main contents include the following: Breeding innovations—This review outlines the taxonomy of the Phalaenopsis genus and highlights its intergeneric hybridization potential, which offers vast opportunities for developing novel horticultural varieties. By establishing clear breeding objectives, researchers employ diverse breeding strategies, including conventional crossbreeding and biotechnological approaches (e.g., mutation breeding, ploidy manipulation, genetic transformation, and CRISPR/Cas9 editing). Propagation and cultivation management—Analyses of Phalaenopsis tissue culture protocols covering explant selection, media optimization, and regeneration systems are summarized. Key factors for efficient cultivation are discussed, including temperature, light, water, nutrient management, cultivation medium selection, and integrated pest/disease management. Scientific environmental control ensures robust plant growth, synchronized flowering, and high-quality flower production. Emerging applications—Phalaenopsis exhibits promising applications in functional bioactive compound extraction (e.g., antioxidants and antimicrobial agents). This review summarizes current advancements in Phalaenopsis breeding, cultivation, and potential applications. Based on technological progress and market demands, future research directions are proposed to support the sustainable development of the Phalaenopsis industry. Full article
(This article belongs to the Special Issue Ornamental Plants and Urban Gardening II)
Show Figures

Figure 1

Back to TopTop