Biology and Life Sciences

Sort by

Review
Biology and Life Sciences
Immunology and Microbiology

Nidhi Satishkumar

,

Som S. Chatterjee

Abstract: Staphylococcus aureus remains to be one of the leading causes of global mortality. The most common class of antibiotics used to treat S. aureus infections are Next-Generation β-lactams (NGBs), as they are highly efficacious and have low adverse effects. NGB resistance in S. aureus is classically attributed to Penicillin-Binding Protein-2a (PBP2a), but previous studies from our group have also implicated altered expression of Penicillin-Binding Protein-4 (PBP4) with high-level NGB resistance. PBP4 is the only low-molecular mass (LMM) PBP present in S. aureus; it is also the only known LMM PBP with transpeptidase activity, giving it the unique ability to bring about peptidoglycan cross-linking. In this article, we review some of the recent findings from our group, which reveal that mutations associated with PBP4 lead to altered protein expression and NGB resistance in both MSSA and MRSA backgrounds. We discuss the clinical relevance of PBP4-associated mutations, particularly in Methicillin Resistant Lacking mec (MRLM) isolates, as well as the combined effect of altered expression of PBP4 and GdpP. Finally, this review summarizes the potential role played by PBP4 in S. aureus virulence. Together, we highlight the increasing relevance of PBP4 as a mediator of NGB resistance and discuss its potential to be an important factor during infection diagnosis and therapy.

Article
Biology and Life Sciences
Immunology and Microbiology

Anna J Harte

,

Elias Mafuru

,

Athumani Ramadhani

,

Tamsyn Derrick

,

Harry Pickering

,

Tara Mtuy

,

Patrick Massae

,

Ehsan Ghasemian

,

Aiweda Malissa

,

Robin L. Bailey

+3 authors

Abstract: Trachoma, caused by Chlamydia trachomatis (Ct), persists as a major cause of preventable blindness despite the global SAFE strategy. Understanding how Ct genovars and genovariants influence infection dynamics and clinical outcomes is crucial for sustaining elimination efforts and informing vaccine development. A four-year longitudinal study was conducted in a trachoma-endemic region of Tanzania across multiple rounds of mass drug administration (MDA) with azithromycin. Ct infections were genotyped by ompA sequencing to identify genovars and genovariants. Associations between genetic variants, bacterial load, and clinical signs of trachoma were assessed. Following MDA, a shift in Ct genovar prevalence occurred from genovar B to genovar A. Genovar B was associated with more severe clinical signs, including follicles, papillae, and scarring, whereas genovar A infections exhibited higher bacterial loads. Among 121 individuals with recurrent infections, 94% were re-infected with the same genovar, indicating limited protective immunity and incomplete clearance despite MDA coverage exceeding 60%. The genovariants B2, B9, and A2 predominated, with an A→T amino acid substitution in B9 potentially modifying antigenic recognition. Post-MDA, normalized genovariant diversity increased, suggesting ongoing transmission or strain reintroduction. Distinct genovar-associated clinical and immunological patterns underscore the need to elucidate genovar-specific virulence and immune evasion mechanisms. These findings provide key insights for optimizing trachoma control and advancing vaccine development.

Article
Biology and Life Sciences
Immunology and Microbiology

Javier Rodríguez López

,

Rosario Lucas López

,

María José Grande

,

Antonio Gálvez

,

Rubén Pérez Pulido

Abstract: A commercial refrigerated vegetable cream containing pumpkin and carrots as main ingredients was stored under refrigeration for 30 days without treatment (controls), supplemented with bacteriocin AS-48, treated by high hydrostatic pressure (HHP, 600 MPa, 8 min, 55ºC) or a combination of bacteriocin and HHP. At day 2, half of the samples were incubated for 24 h at room temperature (simulating a temperature abuse event) and then refrigerated again. Total viable counts and bacterial diversity were determined. Bacteriocin did reduce viable counts, but HHP treatment (singly or in combination with bacteriocin) was the most effective. Viable counts increased in controls during temperature abuse, but not in samples treated with bacteriocin, HHP or both. The initial microbiota of control samples was composed mainly by Pseudomonadota (74.50%), followed by Bacillota (21.19%) and Actinobacteriota (3.69%). Bacillota became the predominant group during refrigerated storage (87.21 to 99.48%). After simulation of a 24-h temperature abuse event, control samples had lower relative abundances of Bacillota during storage and higher relative abundances of Pseudomonadota, Bacteroidota and Actinobacteriota. All treated samples (irrespective of the type of treat-ment) showed a lower relative abundance of Bacillota during storage compared to untreated controls without temperature abuse. Genus Bacillus was the predominant group in the control samples during storage and, although in lower abundance, it was also detected in samples treated with high pressure, bacteriocin or their combination. Acinetobacter was associated with temperature abuse.

Article
Biology and Life Sciences
Immunology and Microbiology

David Vang

,

Pedro Henrique Carneiro

,

Laura Henao

,

Adrien Stroumza

,

Harmony Matshik Dakafay

,

Scott Davis

,

David M. Ojcius

,

Cassio Luiz Coutinho Almeida-da-Silva

,

Aline Cristina de Abreu Moreira-Souza

Abstract:

Porphyromonas gingivalis is a keystone pathogen in periodontitis, known for its ability to invade gingival epithelial cells and persist intracellularly. Conventional antimicrobials are often ineffective against intracellular pathogens, and natural products remain poorly explored in this context. Here, we investigated the antimicrobial effects of Boswellia serrata extract and its bioactive compounds on the dynamics of P. gingivalis infection in human gingival epithelial cells. During early times of infection, B. serrata extracts stimulate endocytic mechanisms and increased bacterial internalization, suggesting a modulation of epithelial uptake mechanisms. At later times of infection, B. serrata increased production of reactive oxygen species (ROS) in host cells and markedly reduced intracellular bacterial load. The antimicrobial effect was abolished by the ROS scavenger N-acetylcysteine, confirming a role for oxidative mechanisms in the clearance of P. gingivalis. Similar results were obtained with 3-O-acetyl-11-keto-β-boswellic acid (AKBA), one of the major boswellic acid derivatives found in B. serrata extract. These findings reveal a dual role of B. serrata compounds in response to P. gingivalis infection, in which B. serrata initially facilitates bacterial entry and subsequently promotes ROS-dependent intracellular killing. These findings provide new mechanistic insights into the regulation of host-pathogen interactions by the natural products found in B. serrata. Our results support the therapeutic potential of B. serrata–derived compounds for managing periodontal infections.

Article
Biology and Life Sciences
Immunology and Microbiology

Caterina Nardella

,

Irene Mezzani

,

Eleonora Pace

,

Alessandra Fierabracci

Abstract: Central tolerance is provided by the AIRE-expressing medullary thymic epithelial cells, through high avidity recognition of self-antigens. Nevertheless, peripheral mechanisms regulate adaptive immunity by deleting autoreactive T-cells that escape thymic selection or inducing their functional unresponsiveness. These mechanisms require interaction with antigen presenting cells exposing cognate antigen. As regard multiple types of extrathymic AIRE-expressing cells, residing in secondary lymphoid organs, were described. In this study we aimed to provide evidence for AIRE binding to promoter regions of known autoantigens in human peripheral blood mononuclear cells (PBMC) in an attempt to elucidate whether this non-classical transcriptional factor could play a role in the pe-ripheral expression of self-antigens. Chromatin immunoprecipitation (ChIP) of 4 normal human PBMC samples was performed using anti-AIRE monoclonal antibody. Quantitative-Real-Time PCR (qRT-PCR) was used to detect AIRE-binding at promoters of known autoantigens, including thyroidrelated thyroglobulin, thyroperoxidase, thyrotropin-receptor, Type 1 diabetes-related autoantigens, i.e. insulin and zinc transporter 8, and to confirm their expression in PBMC. ChIP evidenced amplicons of promoter regions of mentioned autoantigens by qRT-PCR. Expression of AIRE and of autoantigens was confirmed in the same human PBMC samples. This study provides the first evidence that AIRE binds promoters of known autoantigens in human PBMC, supporting its expression and potential role in modulating peripheral self-antigen expression.

Article
Biology and Life Sciences
Immunology and Microbiology

Nay Myo Aung

,

Kyaw Myo Htut

,

Zaw Min Htike

,

Kyaw Khine Win

,

Win Myat

,

Khine Zaw Oo

,

Kyaw Wunna

,

Khine Khine Su

,

Thet Aung

,

Zaw Lwin

Abstract:

Background: The global rise of Multidrug-Resistant (MDR) Escherichia coli (E. coli) represents a critical public health threat, severely compromising the treatment of infections. While Sequence Type 224 (ST224) is recognized as an emergent, high-risk lineage associated with extra-intestinal pathogenic E. coli (ExPEC) and MDR phenotypes globally, its specific genomic features and epidemiological footprint in Southeast Asia, particularly Myanmar, remain largely underexplored. Given Myanmar's vulnerability as an AMR hotspot, comprehensive genomic surveillance is critically leveraged. Method: A laboratory based cross sectional descriptive study conducted at Defense Services Medical Research Centre (DSMRC) during 20th January to 11th November 2025 and aimed to develop Oxford Nanopore Technologies (ONT) long-read sequencing, an early manifestation of this approach for bacterial genomic characterization in Myanmar. Five clinical MDR E. coli isolates (NMA_MM001) from (No.1) Defense Service General Hospital (DSGH) which were identified by Vitek2 analyzer were collected. Extracted DNA was sequenced on the MinION device at DSMRC. Bioinformatic analysis utilized the ONT EPI2ME platform for de novo assembly, followed by MLST, ResFinder, and PlasmidFinder analyses to characterize the isolate's resistome, mobilome, and virulence. Results: Out of five isolates, MDR E. coli (NMA_MM001) of ONT sequencing successfully generated a high-quality, near-closed assembly (N50: 4,911,841 bp, 5 contigs). MLST classified the isolate as ST224. This study confirmed a severe MDR phenotype, identifying blaDHA-1 (AmpC beta-lactamase), blaTEM-1, and two plasmid-mediated quinolone resistance genes (qepA4 and qnrB4). Crucially, the carbapenemase gene blaNDM-5 was identified, located on a highly mobile IncFII plasmid (pAMA1167-NDM-5). This constitutes the first report detailing the emergence of this NDM-5-producing ST224 lineage and its high genomic complexity in Myanmar. Conclusion: This study validates ONT long-read sequencing as an indispensable tool for resolving complex MDR genomes in resource-limited settings. The findings confirm the establishment of an MDR E. coli ST224 isolate in Myanmar carrying the critical blaNDM-5 carbapenemase gene on a highly mobile IncFII plasmid. This genomic information, identification of E. coli ST224, provides an urgent early warning of a highly resistant pathogen, mandating the immediate implementation of targeted infection control measures and regional One Health surveillance programs.

Article
Biology and Life Sciences
Immunology and Microbiology

Simiao Cao

,

Guoxuan Bai

,

Qimuge Wuri

,

Jiayin Li

,

Xiaojing Zhang

,

Zhilin Han

,

Hui Wu

,

Jiaxin Wu

,

Chu Wang

,

Xianghui Yu

+1 authors

Abstract: DNA-based cancer vaccines represent a safe and promising immunotherapeutic strategy, but their clinical efficacy is often limited by weak immunogenicity, primarily due to inefficient antigen cross-presentation. To overcome this challenge, the MHC class I trafficking domain (MITD) can be fused to tumor antigens to enhance their intracellular routing in dendritic cells (DCs), thereby promoting the efficiency of cross-presentation. In addition, incorporation of CD4⁺ T cell epitopes, such as PADRE or P2P16, can robustly activate CD4⁺ T cells, further amplifying antitumor immunity. Thus, combining MITD with CD4⁺ epitopes is expected to synergistically improve DNA vaccine potency. Mesothelin (MSLN), a tumor-associated antigen highly expressed in pancreatic cancer, was selected as the target in this study. We designed MSLN-targeted DNA vaccines incorporating MITD together with either PADRE or P2P16. In a Panc02 murine model, the MITD–PADRE construct, a novel design, elicited stronger immune responses and more effective antitumor activity compared to other formulations. To further counteract immunosuppression, we combined the vaccine with gemcitabine, which enhanced therapeutic efficacy. Together, these findings demonstrate that integrating PADRE with MITD in MSLN-targeted DNA vaccines offers a promising combinatorial strategy for advancing pancreatic cancer immunotherapy.

Review
Biology and Life Sciences
Immunology and Microbiology

Philip Boakye Bonsu

,

Kwadwo Fosu

,

Samuel Badu Nyarko

Abstract: The complications that arise from pregnancy such as preterm birth (PTB), preeclampsia, and neonatal sepsis continue to pose significant threats to maternal and neonatal health worldwide. Early detection and treatment are crucial in reducing the morbidity and mortality of the complications. The human microbiome, particularly during the perinatal period, has also been seen as a critical regulator of immune and metabolic health, influencing both maternal and infant outcomes. This narrative current review responds to the application of microbiome signatures as predictors for maternal and neonatal health biomarkers. We summarize new evidence linking dysbiosis of both maternal gut and vaginal microbiomes with PTB, preeclampsia, and gestational diabetes mellitus (GDM). We further discuss how the neonatal microbiome affects immune development and what is linked to sepsis risk. The review also briefly addresses the crossroads of multi-omics data to enhance precision medicine, the shortcomings in designing adequately powered clinical trials, and the standardization and regulatory hurdles confronting the microbiome field.As much as the potential of microbiome signatures for predictive diagnostics in maternal and neonatal health is high, there are daunting challenges to overcome. They include the dynamic nature of the neonatal microbiome, the nature of multi-omics data complexity, and invoking standardized methodologies and robust clinical trials. Nevertheless, the incorporation of microbiome-based biomarkers into medicine has the potential to move towards more personalized, non-invasive, and effective management of maternal and neonatal health.

Review
Biology and Life Sciences
Immunology and Microbiology

Philip Boakye Bonsu

,

Kwadwo Fosu

,

Samuel Badu Nyarko

Abstract: The tumor microenvironment is a complex environment with interdependence on the relationship between host cells and pathogenic microorganism. This relationship either suppress or promote tumor progression. Pathogens such as H.pylori, F. nucleatum Epstein-Barr virus, Human Papilloma virus and Candida albicans contribute to tumorigenesis through diverse means such as immune checkpoint evasion, genomic instability, reactive oxygen species, viral integration and metabolic reprogramming that favors immunosuppression.Multiomics technologies are relevant in revealing unique host-pathogen signatures that correlate tumor type, tumor staging and therapy response. Artificial intelligence and machine learning models have enabled the integration of genomic, transcriptomic and proteomic and microbiome data to identify pathogen-driven molecular patterns associated with cancer and treatment outcomes. However, translating these findings into clinical practice faces challenges, such as inter-patient variability in microbial composition, the need for external validation across diverse cohorts and the development of standardized cost-effective diagnostic platforms. This narrative review synthesizes current knowledge on the transformative potential of computational frameworks that are available to study these interactions between microbes in the tumor microenvironment and outlines future directions. By bridging the molecular mechanisms with computational innovation, this review provides a roadmap for leveraging hos-pathogen interactions to improve cancer diagnosis, treatment options and patient outcomes.

Review
Biology and Life Sciences
Immunology and Microbiology

Emily Yang

Abstract: The frontlines of innate antiviral immunity center on type I interferons (IFN), which are expressed by nearly all cell types as a cellular alarm signal. IFNs drive the expression of IFN-stimulated genes (ISGs), which can both generate an intracellular antiviral state and regulate the IFN response itself. This key antiviral line of defense is con-served in all jawed vertebrates, including teleost fish. Since their identification nearly 70 years ago, many mammalian ISGs have been identified and characterized However, fish ISGs represent an exciting, largely unexplored avenue of antiviral effector research and present an opportunity to assess how IFN systems have been shaped by whole genome duplication events. This review summarizes advances in identification of bona fide teleost ISGs and examines studies in elucidating the antiviral mechanisms of con-served ISGs, including IFIT1, Mx, Nmi and IFP35, Viperin, TRIMs, and ISG15. Teleost-specific gene expansions and isoform divergence, particularly in the development of the fish novel TRIM family, will be considered under each relevant ISG. Under-standing teleost ISG biology promises not only to improve antiviral strategies in aquaculture but also to reveal novel antiviral principles with translational relevance for human health.

Hypothesis
Biology and Life Sciences
Immunology and Microbiology

Jim L. A. G. van der Zon

,

Frederik P. L. van Loon

Abstract: Psoriasis is traditionally defined and treated as a chronic inflammatory immune-mediated disease. In this manuscript, we propose an alternative, integrative model in which psoriasis isprimarily a disorder of surface and border tissues driven by disturbed cellular metabolism,with inflammation representing a secondary phenomenon. Psoriasis predominantly manifestsin anatomical border zones, such as skin folds, hairlines, nail beds, and joint surfaces,wherecells depend critically on diffusion- and osmosis-based metabolic processes. These vulnerableregions are highly sensitive to alterations in microcirculation, oxygen delivery, nutrientavailability, and blood composition.We argue that a range of comorbidities and risk factors, including thyroid dysfunction,obesity, hyperhomocysteinemia, smoking, nutritional deficiencies (notably vitamin D and zinc), stress-related vasospasm, and dietary imbalances, may impair mitochondrial aerobicmetabolism in these border cells. This metabolic disruption promotes a shift from aerobicrespiration to anaerobic glycolysis, leading to the accumulation of mitochondrial wasteproducts and crystalline debris, particularly in confined tissue spaces such as joints. We hypothesize that this metabolic debris acts as the primary trigger for the characteristicinflammatory response observed in psoriasis.The dual metabolic and clinical efficacy of fumarates provides key support for this model, asthese agents both restore mitochondrial metabolic balance and alleviate psoriatic inflammation. This observation suggests that inflammation is a downstream consequence ofmetabolic failure rather than the initiating cause of disease. Based on this framework, wepropose that therapeutic strategies should prioritize correction of metabolic dysfunction and associated comorbidities, after which inflammatory manifestations are expected to diminish. Such an approach may not only improve cutaneous and articular symptoms but also reduce psychological burden and social stigma associated with psoriasis.

Article
Biology and Life Sciences
Immunology and Microbiology

Ofélia Godinho

,

Olga Maria Lage

,

Sandra Quinteira

Abstract:

Background/Objectives: It is of upmost importance to study environmental bacteria, as these microorganisms remain poorly characterized regarding their diversity, antimicrobial resistance, and impact on the global ecosystem. This knowledge gap is particularly pronounced for marine bacteria. In this study, we aimed to isolate marine bacteria from different sources and to gain insights into the environmental bacterial resistome, an aspect that remains largely neglected. Methods: Bacteria were isolated from several marine sources using two different culture media, and their identification was based on 16S rRNA gene analysis. Whole-genome sequencing was performed for selected isolates belonging to novel taxa. Antimicrobial susceptibility to seven antibiotics was evaluated using the disk diffusion method. Results: A total of 171 bacterial isolates belonging to the phyla Pseudomonadota, Bacteroidota, Planctomycetota, Actinomycetota, and Bacillota were obtained from diverse marine samples. The most abundant group belonged to the class Alphaproteobacteria. Thirty isolates represented novel taxa, comprising 16 new species and one new genus. Despite the challenges associated with determining antibiotic resistance profiles in environmental bacteria, only one isolate (1.8%) was pan-susceptible, whereas 54 (98.2%) showed resistance to at least one of the tested antibiotics. Moreover, 33 isolates exhibited a multidrug-resistant phenotype. Genome analysis of four novel taxa revealed the presence of an incomplete AdeFGH efflux pump. Conclusions: This study highlights the high bacterial diversity in marine environments, the striking prevalence of antibiotic resistance, and the major methodological challenges in studying environmental bacteria. Importantly, it emphasizes the relevance of culturomics-based approaches for uncovering hidden microbial diversity and characterizing environmental resistomes.

Review
Biology and Life Sciences
Immunology and Microbiology

Andreea Laura Antohi

,

Andreea Daria Gheorghiță

,

Octavian Andronic

,

Gratiela Gradisteanu Pircalabioru

,

Andreea-Ramona Treteanu

Abstract: Recent findings suggest that the gut microbiome significantly influences cancer outcomes, including responses to immune checkpoint inhibitor (ICI) treatments. Although early research focused on gut bacteria, it is now understood that the microbiome includes a bacteriome, virome, and mycobiome, all of which can modulate host immunity. Some commensal bacteria enhance anti-tumor immune responses and improve ICI efficacy, as demonstrated in both mice and patients. Fecal microbiota transplants (FMT) from patients responding to ICI have successfully reversed resistance in certain non-responders. In addition to bacteria, gut fungi and viruses are gaining attention as further factors influencing ICI effiectiveness and toxicity. Recent multi-omics studies across cancer cohorts show that fungal and viral populations in the gut vary between ICI responders and non-responders. Commensal fungi may shape anti-cancer immunity by inducing inflammatory or tolerogenic pathways, while viral components can stimulate innate immune sensors that promote tumor surveillance. On the other hand, gut dysbiosis marked by expansion of pathobionts (including opportunistic fungi) and reduction of beneficial microbes is linked to serious immune-related adverse events (irAEs) such as ICI-induced colitis. This review discusses the multi-kingdom gut microbiome – bacteria, fungi, and viruses – and their interactions with the immune system in cancer therapy. We emphasize known mechanisms linking these microbes to anti-tumor immunity, overview human studies associating gut microbiome profiles with ICI outcomes and explore strategies to modulate the microbiome to enhance ICI efficacy while reducing toxicity. Understanding and utilizing the gut mycobiome and virome in conjunction with the bacteriome could pave the way for new biomarkers and therapeutic adjuvants in cancer immunotherapy.

Review
Biology and Life Sciences
Immunology and Microbiology

Alexander Jacob

,

Harbir Singh

,

Satheesh Kumar Poolakkad Sankaran

,

Rahul K. Das

,

Ravi Aalinkeel

,

Supriya D. Mahajan

,

Jessy J. Alexander

Abstract: The complement system is a central component of innate immunity with established roles in host defense and emerging functions in neurodevelopment, synaptic remodeling, and neuroimmune communication within the central nervous system (CNS). In parallel, advances in nanotechnology have enabled targeted strategies for CNS drug delivery but have also revealed that many nanomaterials interact with and activate complement, influencing biodistribution, safety, and inflammatory responses. Opioid use disorder (OUD) is increasingly recognized as a condition associated with chronic neuroimmune dysregulation involving glial activation, altered cytokine signaling, and blood-brain barrier (BBB) disruption. Although relatively few studies have directly measured complement activation in OUD, emerging transcriptomic, cellular, and inflammatory data suggest that complement pathways may intersect with opioid-induced neuroimmune signaling. This review synthesizes current knowledge at the intersection of complement biology, nanomedicine, and opioid-associated neuroimmune changes. It distinguishes well-established mechanisms of complement activation by nanomaterials from emerging evidence linking complement signaling to opioid exposure. It integrates complement pathways with opioid receptor and Toll-like receptor 4 (TLR4) signaling in glial cells and endothelial compartments, and discusses both beneficial and pathological roles of complement in the CNS. Finally, the therapeutic potential and limitations of complement-aware nanotechnology and complement modulation in CNS drug delivery and addiction neuroscience are outlined to guide translation of complement-targeted nanomedicines in addiction neuroscience.

Article
Biology and Life Sciences
Immunology and Microbiology

Jiani Liu

,

Destin T. Hinson

,

Michael Hansen

,

Virginia P. Van Keulen

,

Brian J. Parrett

,

Larry Pease

,

Michael A. Barry

Abstract:

Background/Objectives: Antigen presenting cells (APCs) and immune cells have unique properties to drive or suppress immune responses. They are therefore key targets for the expression of vaccine antigens or transgene proteins. To better determine the utility of different molecular therapies to modify these cells, mRNA and DNA-based molecular therapy vectors were compared for their ability to genetically modify immune cells after intradermal injections in mice. DNA-based vectors included naked plasmid DNA, plasmid packaged in lipid nanoparticles (LNPs), and replication-defective adenovirus (Ad) vectors. mRNA delivery was mediated by packaging into LNPs like those used in COVID-19 vaccines. Methods: Each vector was used to deliver Cre recombinase into Cre reporter mice whose cells are activated to express green fluorescent protein (GFP) and firefly luciferase after Cre recombination. Mice were injected intradermally (ID) near the base of their tail at a site that drains into the inguinal lymph node. Luciferase activity was imaged in the living mice 1 or 4 days after vector injection. The animals were then euthanized and luciferase activity was imaged in the draining inguinal lymph node. Cells were prepared from the intradermal injection site and from the draining lymph node to determine which immune cells were genetically modified by phenotyping CD45, CD3, and CD11b GFP-positive cells by flow cytometry. Given that the skin uniquely contains Langerhans dendritic cells, these CD207+ cells were also phenotyped in skin samples and in the draining lymph node. Results: In both the skin and in the draining lymph node, the rank order of luciferase and GFP activation by the vectors were: 1) Ad; 2) mRNA-LNP; 3) DNA-LNP; and 4) naked DNA. Only mRNA-LNP and Ad vectors mediated obvious luciferase activity in the living animals and in the draining lymph nodes by imaging. Notably, both vectors appeared to leak from the ID injection site and not only modify the draining lymph node but also strongly modify the livers of the mice. Naked DNA and DNA-LNP mediated detectable GFP activation in the skin and draining lymph node in some mice, but this activity was low and did not reach statistical significance when compared to PBS-treated animals. mRNA-LNPs and Ad both mediated significant Cre delivery in CD45+, CD3+, CD11b+, and CD207+ immune cells in the skin and in the lymph node with adenovirus mediating consistently higher levels of expression in all of the tested cells. Conclusions: These data indicate that mRNA-LNP and Ad vectors mediate stronger modification of skin and lymph node immune cells after intradermal injections. Naked DNA and DNA-LNPs were markedly less potent at this activity than the other vectors. These data are consistent with the higher vaccine potency of mRNA-LNP and Ad vectors and suggest that approaches that increase targeting of immune cell subsets may have utility to increase efficacy while also reducing off target modification of tissues like the liver.

Article
Biology and Life Sciences
Immunology and Microbiology

Praveen Kumar

,

Mohit Kumar

,

Amandeep Saini

,

Sheikh Owais Mohamad

,

Basharat Ali

,

Brooke D. Esquivel

,

Atanu Banerjee

,

Theodore C White

,

Naseem A Gaur

,

Abdul Haseeb Shah

+2 authors

Abstract: In Candida species, including Candidozyma auris (formerly Candida auris), overexpression of efflux pumps is a well-established mechanism of antifungal resistance. However, accumulating evidence indicates that impaired drug import may also significantly contribute to reduced antifungal susceptibility. Sugar importers, historically viewed solely as hexose transporters (HGTs), are now emerging as potential indirect modulators of antifungal uptake. Here, we performed a comprehensive inventory and functional analysis of the HGT family in C. auris to assess its contribution to antifungal import. Phylogenetic analyses revealed that C. auris HGTs are more closely related to those of Candida albicans (C. albicans) than Saccharomyces cerevisiae (S. cerevisiae). All HGT genes showed basal expression, with several significantly downregulated upon fluconazole (FLC) exposure. To establish functional relevance, we generated a mini-library of HGT deletion mutants. Notably, the Δhgt13 strain exhibited markedly increased FLC resistance, concomitant with reduced intracellular FLC accumulation and decreased membrane permeability. Consistently, molecular docking and molecular dynamics simulations demonstrated strong and stable interactions between FLC and Hgt13p. Together, these findings implicate Hgt13p as a key determinant of FLC import and membrane permeability, revealing reduced FLC import could also contribute to antifungal resistance in C. auris.

Article
Biology and Life Sciences
Immunology and Microbiology

Pompilio Arianna

,

Di Bonaventura Giovanni

Abstract:

Background/Objectives: Stenotrophomonas maltophilia is an emerging opportunistic pathogen associated with severe infections, particularly in patients with cystic fibrosis (CF). Its intrinsic multidrug resistance and ability to form biofilms significantly complicate treatment. While biofilm growth is widely linked to antimicrobial tolerance, the relationship between biofilm-forming capacity and planktonic antibiotic resistance in S. maltophilia remains unclear. This study aimed to investigate the association between antibiotic resistance profiles and biofilm formation in clinical isolates from CF and non-CF patients. Methods: A total of 86 clinical S. maltophilia isolates (40 from CF airways and 46 from non-CF patients) were analyzed. Antibiotic susceptibility to seven agents was assessed by disk diffusion, with results interpreted according to EUCAST and CLSI criteria. Multidrug resistance phenotypes were defined using standard criteria. Biofilm formation was quantified after 24 h using a crystal violet microtiter plate assay and categorized into five levels of production. Statistical analyses were performed to compare biofilm formation across resistance profiles and clinical origins and to assess correlations between biofilm biomass and multidrug resistance. Results: Overall, high resistance rates were observed, particularly to meropenem (87.2%), ciprofloxacin (80.2%), and rifampicin (72.1%). CF isolates showed significantly higher resistance to piperacillin/tazobactam and a higher prevalence of multidrug resistance. Biofilm production was detected in 94.2% of isolates, with strong and powerful biofilm producers predominating. However, isolates from CF patients formed significantly less biofilm than those from non-CF patients. Notably, resistance to piperacillin/tazobactam and meropenem was associated with significantly reduced biofilm formation, as reflected in both median biomass and the proportion of high biofilm producers. Across the entire collection, the number of antibiotic resistances displayed by an isolate was negatively correlated with biofilm biomass. These trends were maintained after stratification by clinical origin, although some comparisons did not reach statistical significance. Conclusions: These findings demonstrate an unexpected inverse relationship between planktonic antibiotic resistance and biofilm-forming efficiency in S. maltophilia. Enhanced biofilm production may represent an alternative persistence strategy in more antibiotic-susceptible strains, with important implications for infection management and therapeutic failure.

Review
Biology and Life Sciences
Immunology and Microbiology

Ola A Al-Ewaidat

,

Moawiah M Naffaa

Abstract: Engineered microbes are emerging as a new class of living immunotherapeutics capable of sensing, interpreting, and actively reshaping host immune systems. Unlike conventional biologics or cell therapies with fixed mechanisms of action, engineered microbial platforms operate as dynamic systems that integrate environmental, metabolic, and immunological cues, process these inputs through programmable biological circuits, and execute context-dependent immune modulation with spatial and temporal precision. This review presents an immune-first framework that conceptualizes engineered microbes as distributed immune-computational systems defined by coordinated sensing, signal processing, memory, and effector functions embedded within host immune networks. Organizing the field around immune logic rather than microbial taxonomy or disease category, we examine how engineered microbes detect tissue-specific and immune-state signals, translate these inputs through synthetic processing modules, and generate immune outputs that activate, suppress, educate, or reprogram immunity across cancer, autoimmunity, and infectious disease. We further define immune safety architecture as a core design principle governing inflammatory control, tolerance preservation, adaptive immunity, and therapeutic termination, and discuss the translational and regulatory implications of immune-state–resolved clinical evaluation. Together, this framework positions engineered microbes as programmable immune systems and establishes a unifying conceptual foundation for their development as next-generation living immunotherapies.

Review
Biology and Life Sciences
Immunology and Microbiology

Angel Justiz-Vaillant

Abstract: Chimeric antigen receptor (CAR) immunotherapy has made significant strides, particularly in hematological malignancies, with five CAR T therapies gaining FDA approval. However, its application in solid tumors remains challenging due to issues like poor CAR T cell trafficking, an immunosuppressive tumor microenvironment (TME), and therapy-related toxicity. CAR Natural Killer (NK) cells present an alternative with advantages such as multiple mechanisms for targeting cancer and reduced side effects. Additionally, macrophages, which naturally infiltrate tumors, are under investigation for CAR therapy to overcome the limitations seen in CAR T and CAR NK approaches. In this review, our primary focus will be on solid tumors, as they present unique challenges in CAR T cell therapy, such as poor trafficking and the immunosuppressive tumor microenvironment. However, we will also address some hematological malignancies, particularly those with poor prognoses, where similar issues of CAR T cell dysfunction and lack of persistence are observed. By covering both solid tumors and certain blood cancers, we aim to provide a comprehensive understanding of the barriers and potential strategies for improving CAR T cell therapies across different malignancies.

Review
Biology and Life Sciences
Immunology and Microbiology

Umama Shahid

Abstract: Antimicrobial resistance (AMR) is increasingly addressed through genomic approaches that identify resistance genes and mutations. While these methods have improved surveillance and diagnostics, they often fail to explain patient-specific treatment outcomes, as genetically similar pathogens can exhibit markedly different responses to the same antimicrobial therapy. This discrepancy highlights a fundamental limitation of gene-centric frameworks: resistance is not solely a static genetic property, but a dynamic physiological state shaped by regulatory, metabolic, and environmental factors. This review synthesizes current evidence supporting a transcriptomics-driven perspective of AMR, in which resistance is conceptualized as a context-dependent “resistance state” emerging from regulated gene expression. Pathogen transcriptomics captures functional activity that is invisible to genomic data alone, revealing how transcriptional programs underlying tolerance, persistence, inducible efflux, and stress adaptation contribute to antimicrobial survival without stable genetic change. Experimental and host-relevant studies demonstrate that these transcriptional states are strongly modulated by antibiotic exposure, host immune pressures, infection site physiology, and microbiome context, providing a mechanistic basis for inter-patient variability in treatment response. The review critically examines recent efforts to develop expression-based resistance signatures and discusses the opportunities and limitations of integrating transcriptomics into precision AMR diagnostics. Emphasis is placed on validation requirements, interpretability, and clinical feasibility, as well as on the importance of outcome-linked evidence. Finally, key knowledge gaps and future directions are outlined, including the need for standardized resistance-state definitions, physiologically relevant models, and multicentre clinical validation. By reframing AMR as a dynamic and measurable resistance state, transcriptomics offers a complementary layer to existing diagnostics and a potential pathway toward more precise, individualized antimicrobial therapy.

of 92

Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated