Microalgae are a group of photosynthetic autotrophic microorganisms that are classified as Generally Recognized as safe (GRAS). They are rich in high-value bioactive compounds with broad applications in food, healthcare and pharmaceuticals. Recent research demonstrated that microalgae have significant potential as innovative biomaterials for biomedical applications. The unique phototactic movement of microalgae enables them controlled drug delivery to targeted tissues in patients. Furthermore, microalgae produce oxygen via photosynthesis when exposed to light, overcoming tumor hypoxia limitations and improving biomedical imaging in vivo. Additionally, the intrinsic biophysical properties and modifiability of microalgae can be harnessed for the development of biohybrid robots and bioprinting, expanding their clinical applications. This review highlights current engineering innovations in microalgae for medical applications, such as drug delivery, tumor hypoxia targeting, wound healing, and immunotherapy. The remarkable biocompatibility, diverse biological functionalities, and cost-effectiveness of microalgae provide a promising platform for future application of targeted drug delivery and precision medicine.
In recent years, single-cell omics technologies have significantly advanced plant and agricultural research, providing transformative insights into plant development, cellular heterogeneity, and environmental response mechanisms. Traditional bulk-level analyses often obscure differences between individual cells, whereas single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq) now reveal unique expression profiles across distinct cell populations, facilitating the identification of novel cell types and elucidation of gene regulatory networks. Additionally, epigenomic approaches like single-nucleus ATAC sequencing (snATAC-seq) offer a deeper understanding of chromatin accessibility and its complex relationship with gene regulation. These technologies have seen widespread application in model plants such as Arabidopsis thaliana, as well as in major crops and horticultural plants, providing essential data for crop improvement and breeding strategies. Moving forward, with the continued development and integration of single-cell multi-omics technologies, there will be greater depth of insight into cell-type-specific regulation and complex trait analysis, bringing new opportunities for sustainable agriculture and crop improvement.
The growth and adaptability of desert plants depend on their rhizosphere microbes, which consist of a few abundant taxa and numerically dominant rare taxa. However, the differences in diversity, community structure, and functions of abundant and rare taxa in the rhizosphere microbiome of the same plant in different environments remain unclear. This study focuses on the rhizosphere microbial communities of Artemisia desertorum, a quintessential desert sand-stabilizing plant, investigating the diversity patterns and assembly processes of rare and abundant taxa across four Chinese deserts: Mu Us, Kubuqi, Tengger, and Ulan Buh. The results show that climatic factors, especially aridity and mean annual precipitation (MAP), significantly influence bacterial community composition and microbial network complexity. The interactions between rare and non-rare taxa are non-random, forming a modular network in which rare taxa serve as central nodes, and their loss could destabilize the network. Rare taxa are primarily shaped by heterogeneous selection, whereas abundant taxa are mainly influenced by dispersal limitation. Functionally, abundant taxa exhibit higher metabolic potential, whereas rare taxa are more involved in processes such as cell motility, indicating distinct ecological roles. These results provide new insights into the ecological functions of rare and abundant taxa in desert rhizosphere communities and highlight the importance of microbial management for desert plant health.
Arbuscular mycorrhizal fungi (AMF) have the potential to enhance plant tolerance to abiotic stresses. However, the impact of AMF on the rhizosphere bacterial community of tobacco under conditions of low nutrient availability remains unclear. This study investigated the influence of inoculating Claroideoglomus etunicatum on the tobacco rhizosphere bacterial community and the microbial mechanisms by which AMF enhanced plants antioxidant capacity, employing Illumina MiSeq high-throughput sequencing. The findings indicated that AMF significantly increased both the aboveground and belowground fresh weight, as well as the plant height of tobacco. AMF inoculation led to elevated activities of catalase (CAT) and superoxide dismutase (SOD), a reduction in malondialdehyde (MDA) content, and an overall enhancement of the plants antioxidant capacity. Phylogenetic analysis demonstrated that AMF modified the bacterial community structure and significantly enriched beneficial rhizosphere bacteria, predominantly from the phyla Proteobacteria, Chloroflexi, Actinobacteriota, and Myxococcota, thereby facilitating tobacco growth. The network analysis revealed that the incorporation of arbuscular mycorrhizal fungi (AMF) contributed to increased stability within the bacterial community, enriched species diversity, and more intricate ecological networks. AMF enhanced interactions and positive correlations among bacterial species, indicating that heightened microbial synergy is associated with improved symbiotic relationships. Furthermore, the structural equation model demonstrated that AMF bolstered the plants antioxidant capacity by modulating the rhizosphere bacterial community. This study elucidates the impact of AMF on the tobacco rhizosphere bacterial community, providing a theoretical basis for promoting tobacco growth.
The mannose receptor (MR) is a member of the C-type lectin superfamily and a type I transmembrane protein that functions as a pattern recognition receptor (PRR) in immune responses. In this study, we identified 13 MR genes (RpMR1-13) in the genome of Ruditapes philippinarum and investigated their expression profiles following Vibrio anguillarum challenge. Notably, RpMR1, RpMR2, RpMR3, and RpMR4 exhibited peak expression at 72 h post-infection. We successfully purified the recombinant RpMR1 protein and demonstrated its antibacterial activity against three Gram-negative bacteria (V. splendidus, V. alginolyticus, and V. anguillarum), though it had no effect on Gram-positive bacteria. Furthermore, in vivo injection of RpMR1 significantly reduced mortality in R. philippinarum following V. anguillarum infection. To explore role of RpMR1 in immune signaling, we performed RNA interference (dsRNA-RpMR1) and observed successful gene silencing. Subsequent qRT-PCR analysis revealed that RpMR1 knockdown significantly suppressed TLR4 expression (P< 0.05) under V. anguillarum stress, confirming an interaction between RpMR1 and TLR4 in the immune response. This study provides the first functional evidence of mannose receptor-mediated immunity in mollusks, offering new insights into the molecular defense mechanisms of R. philippinarum against bacterial infection.
Approximately 75% of the human genome is transcribed into RNA, yet less than 5% encodes proteins, with the majority producing non-coding RNAs (ncRNAs). Among them, long non-coding RNAs (lncRNAs) represent a major class that exerts broad regulatory influence across cellular processes, disease contexts, and developmental stages. Despite their potential as biomarkers and therapeutic targets, their low sequence conservation, limited abundance, and structural complexity present significant challenges for functional characterization. Traditional RNA interference and CRISPR-Cas9–based methods have offered partial insights but remain limited in efficiency, specificity, and scalability. To address these barriers, Neville E. Sanjana’s team developed CaRPool-seq, a transcriptome-scale CRISPR-Cas13 screening platform that directly targets RNA. Applying this approach across diverse human cell lines, they identified 778 essential lncRNAs, including 46 universally required for survival, with distinctive structural features and functional independence from neighboring protein-coding genes. Integration with single-cell transcriptomics revealed their critical roles in cell-cycle regulation, apoptosis, and developmental gene expression, as well as aberrant expression patterns in cancer linked to patient outcomes. This study establishes CRISPR-Cas13 as a precise and scalable strategy for lncRNA functional discovery, expanding opportunities for biomarker identification, therapeutic development, and precision medicine.
Trichomes are crucial for plant defense and secondary metabolite biosynthesis. In Artemisia argyi, T-shaped non-glandular trichomes (TSTs) are a defining morphological feature and the primary structural component of moxa floss. We observed pronounced TST accumulation on the lower leaf surfaces. To elucidate the genetic regulation of TST development, we performed comparative transcriptomics of TSTs and non-TST tissues. This identified several MIXTA-like transcription factors (named AarMIXTAs) as key regulators of TST differentiation. Phylogenetic analyses revealed gene expansion and functional divergence between the AarMIXTAs and their homologs in Artemisia annua. Heterologous overexpression of AarMIXTA1.2 in Arabidopsis significantly increased TST density, demonstrating its positive regulatory role via transcriptional activation of downstream targets. These findings elucidate molecular mechanisms underlying TST morphogenesis and provide a genetic framework for enhancing moxa floss yield in A. argyi cultivars.
In plants, autophagy is a conserved recycling system essential for development and stress responses by targeting cellular components for massive degradation in the vacuole. Our previous work suggested that autophagy contributes to Arabidopsis (Arabidopsis thaliana) stress responses by modulating NADPH-oxidase-mediated reactive oxygen species (ROS) homeostasis; however, the molecular link between extracellular ROS and autophagy remains unknown. We performed a yeast two-hybrid screen to identify components involved in autophagy, using the central autophagy component ATG8e as a bait. We identified MEMBRANE ATTACK COMPLEX/PERFORIN-LIKE 2 (MACP2) as an interactor of ATG8e via its the ATG8-interacting motif and confirmed this interaction by co-immunoprecipitation and bimolecular fluorescence complementation assays. MACP2-overexpressing lines showed enhanced sensitivity to nutritional starvation, accelerated leaf senescence, and increased hydrogen peroxide (H2O2) levels, resembling the phenotypes of atg mutants defective in autophagy. Conversely, macp2 knockouts exhibited diminished starvation-induced H2O2 accumulation and attenuated autophagosome formation and fully suppressed the starvation-hypersensitive phenotypes of the atg5-1 mutant. In particular, MACP2 was degraded through the autophagy machinery during prolonged starvation, suggesting a feedback regulatory mechanism for maintaining MACP2 homeostasis. Our findings suggest that MACP2 acts as a key regulator in autophagy induction by controlling influx of extracellular H2O2 in Arabidopsis.