Latest Articles

58 articles
Active filters: Nature Biomedical Engineering ×



N
Nature Biomedical Engineering · Nov 18, 2025

Label-free navigation system for grading prostate tumour malignancy in situ via tissue pH and prostate-specific antigen activity

Radical prostatectomy is a standard curative approach for high-risk prostate cancer, yet accurately defining tumour margins during surgery remains a major challenge. Intraoperative assessment of prostate tumour malignancy—particularly those with high aggressiveness catalogued in Gleason grade group (GG) ≥ 3—is crucial to prevent positive surgical margins and minimize postoperative complications. Here we develop a surface-enhanced Raman scattering (SERS)-based navigation system for intraoperative localization of high-grade malignant regions by simultaneously accessing tissue acidity and prostate-specific antigen (PSA) enzymatic activity. This system integrates a sampling pen for automated biomarker extraction from tissue surfaces, a nano-imprinted SERS array producing a ratiometric Raman signal in response to acidity and PSA activity, and a two-dimensional deep-learning model for rapid Raman spectral interpretation. We show that the system can intraoperatively identify GG ≥ 3 malignancies in fresh prostate tissues from 144 Chinese patients with an area under the receiver operating characteristic curve of 0.89. This SERS-based navigation system holds strong potential to enhance surgical precision, minimize tumour residue and ultimately improve patient outcomes.

Microfluidics Molecular imaging Nanomedicine Prostate Raman spectroscopy



N
Nature Biomedical Engineering · Nov 05, 2025

A pre-trained large generative model for translating single-cell transcriptomes to proteomes

Measuring protein abundance at the single-cell level can facilitate a high-resolution understanding of biological mechanisms in cellular processes and disease progression. However, current single-cell proteomic technologies face challenges such as limited coverage, constrained throughput and sensitivity, batch effects, high costs and stringent experimental operations. Inspired by the translation procedure in both natural language processing and the genetic central dogma, we propose a pre-trained, large generative model named single-cell translator (scTranslator). scTranslator can generate multi-omics data by inferring the missing single-cell proteome based on the transcriptome. Through systematic benchmarking and validation on independent datasets, we have confirmed the accuracy, stability and flexibility of scTranslator across various profiling techniques (for example, CITE-seq, spatial CITE-seq, REAP-seq, NEAT-seq), cell types (for example, monocytes, macrophages, T cells, B cells), tissues (for example, blood, lung, brain) and a wide range of disease contexts, including infectious, metabolic and oncologic conditions. Furthermore, scTranslator shows its superiority in assisting various downstream analyses and applications, including gene/protein interaction inference, perturbation prediction, cell clustering, batch correction and cell origin recognition in pan-cancer data.

Machine learning Proteome informatics


N
Nature Biomedical Engineering · Nov 03, 2025

Nonexpansive biodegradable matrix promotes blood vessel organoid development for neurovascular repair and functional recovery in ischaemic stroke

Tissue engineering-based vascular reconstruction represents a promising therapeutic strategy for ischaemic stroke. However, in the confined stroke cavity, conventional implants are unable to simultaneously provide swelling-resistant support and growth-permissive internal space, which are crucial for effective revascularization. To address this limitation, we develop a bioinspired, non-expansive biodegradable matrix (NEBM) through covalent–non-covalent assembly of commercially available, clinical-grade natural polymers. We show that NEBM recapitulates key features of brain extracellular matrix—including porous microstructure and tissue-matched stiffness—to deliver structural stability. Moreover, its progressively degradable structure establishes a dynamic remodelling niche that directs cellular behaviour towards promoting angiogenesis. Compared with commercial Matrigel-based matrix, NEBM fosters blood vessel organoid development with higher vascular density, larger vessel diameters and more distinct arterial features. In both subcutaneous and stroke transplantation models, we find that NEBM facilitates the integration of blood vessel organoids with the host vasculature. Strikingly, this revascularization in stroke cavity stimulates neurogenesis, contributing to significant functional recovery. As such, our study provides valuable guidance to design clinically translatable matrices for organ repair and regeneration in confined environments.

Bioinspired materials Biomedical engineering Gels and hydrogels Implants

N
Nature Biomedical Engineering · Nov 03, 2025

Engineered human induced pluripotent stem cell models reveal altered podocytogenesis in congenital heart disease-associatedSMAD2mutations

Clinical observations of patients with congenital heart disease carryingSMAD2genetic variants revealed correlations with multi-organ impairments at the developmental and functional levels. Many patients with congenital heart disease present with glomerulosclerosis, periglomerular fibrosis and albuminuria. It remains largely unknown whetherSMAD2variants associated with congenital heart disease can directly alter kidney cell fate, tissue patterning and organ-level function. Here we investigate the role of pathogenicSMAD2variants in podocytogenesis, nephrogenic cell lineage specification and glomerular filtration barrier function using a combination of CRISPR-based disease modelling, stem cell and microfluidic organ-on-a-chip technologies. We show that the abrogation ofSMAD2results in altered patterning of the mesoderm and intermediate mesoderm cell lineages, which give rise to nearly all kidney cell types. Following further differentiation of intermediate mesoderm cells, the mutant podocytes failed to develop arborizations and interdigitations. A reconstituted glomerulus-on-a-chip system showed substantial albumin leakage, as observed in glomerulopathies. This study implicates chronic heart disease-associatedSMAD2mutations in kidney tissue malformation that might inform targeted regenerative therapies.

Biomedical engineering Development Paediatric kidney disease Stem-cell differentiation Urological manifestations

N
Nature Biomedical Engineering · Oct 31, 2025

Systematic production of human kidney organoids for transplantation in porcine kidneys during ex vivo machine perfusion

Organoids derived from human pluripotent stem (hPS) cells hold promise for therapeutic purposes. However, technological advances to overcome their massive production while ensuring differentiation fidelity are still lacking. Here we report a procedure sustaining the derivation of kidney organoids from hPS cells (hPSC-kidney organoids) using a scalable, reproducible and affordable approach that allows hPSC-kidney organoid differentiation into different renal cell types. Using single-cell RNA sequencing, confocal image analysis, metabolic assays and CRISPR–Cas9 engineering for generation of fluorescent reporters, we show that hPSC-kidney organoids exhibit transcriptional variety and cellular composition following cell-to-cell contact. We infuse human kidney organoids into ex vivo porcine kidneys using normothermic machine perfusion, and demonstrate in vivo engraftment of hPSC-kidney organoids. We further evaluate the immune response, confirming the feasibility and viability of the procedure. We identify cells of human origin after normothermic machine perfusion and in vivo transplantation by means of in situ hybridization, immunohistochemistry, confocal microscopy, image analysis and quantification, in vivo imaging, and flow cytometry. This work provides a foundation for using hPSC-kidney organoids for ex vivo cell-based therapies in clinical trials.

Differentiation Induced pluripotent stem cells

N
Nature Biomedical Engineering · Oct 31, 2025

Steric stabilization-independent stealth cloak enables nanoreactors-mediated starvation therapy against refractory cancer

The high interfacial energy of nanomaterials limits their certain biomedical applications that require stealthiness to minimize non-specific interaction with biological components. While steric repulsion-based entropic stabilization—such as PEGylation—has long been the dominant strategy for designing stealth nanomaterials, its inherent softness and susceptibility to dynamic deformation and external forces often result in only moderate stealth performance. Here we report a distinct approach to achieving stealthiness by harnessing an ion-pair network, rather than maximizing steric repulsion. Using model polyion complex nanoparticles composed of equimolar charge ratios of polycations and polyanions, we demonstrate that increasing crosslinks between the constituent polyions beyond a critical threshold effectively reduces protein adsorption and macrophage uptake, enabling prolonged circulation with a half-life exceeding 100 hours. Building on this, we develop an asparaginase-loaded vesicular nanoreactor enveloped by a semi-permeable ion-pair network sheath for asparagine starvation therapy. The extended circulation of these nanoreactors enables sustained depletion of asparagine, leading to improved therapeutic outcomes for metastatic breast and pancreatic cancers. Our findings open an avenue for improving the pharmacokinetics of nanomaterials for therapeutic delivery through delicately engineering stable intermolecular structures with holistic cooperativity.

Cancer immunotherapy Cancer metabolism Drug delivery Metastasis Protein delivery

N
Nature Biomedical Engineering · Oct 30, 2025

High-throughput evaluation of in vitro CRISPR activities enables optimized large-scale multiplex enrichment of rare variants

Previous high-throughput evaluations of CRISPR activities for a large number of target and guide RNA sequences were based on measuring insertion–deletion frequencies rather than cleavage efficiencies. Here we develop two high-throughput in vitro methods, Cut-seq1 and Cut-seq2, to evaluate Cas9 cleavage efficiency for tens of thousands, or even hundreds of thousands, of guide RNA–target pairs. These methods reveal low correlations between in vitro cleavage efficiencies and insertion–deletion frequencies in cells, yet high concordances in protospacer adjacent motif compatibility. Using the resulting large datasets of in vitro cleavage efficiencies, we develop DeepCut, a set of deep learning models that can identify optimized single-guide RNAs that can selectively cleave specific sequences, even in the presence of similar noise sequences. Using these optimized single-guide RNAs, we develop a method, CLOVE-seq (which stands for cleavage for large-scale optimized variant enrichment sequencing), to enrich rare variants in a multiplexed manner by Cas9-mediated specific cleavage of noise or rare variant sequences. Our methods can enhance the understanding of CRISPR nuclease activities and could be used to detect a large number of rare variants in various biomedical contexts.

CRISPR-Cas9 genome editing Genetic engineering High-throughput screening


N
Nature Biomedical Engineering · Oct 29, 2025

A targeted vector for brain endothelial cell gene delivery and cerebrovascular malformation modelling

Defects in brain endothelial cells (brainECs) can cause severe cerebrovascular malformations, including arteriovenous malformation (AVM) and cerebral cavernous malformation. The lack of appropriate tools for cerebrovascular disease modelling and local genetic manipulation of the brain vasculature hinders research on cerebrovascular malformations. Here we develop a recombinant adeno-associated virus (rAAV) tool termed miniBEND (rAAV-based mini-system for brain endothelial cells, rAAV-miniBEND), which combines a minimal promoter and an optimizedcis-acting element isolated from the mouse geneTek. This system activates gene expression specifically in mouse and rat brainECs. rAAV-miniBEND achieved high-efficiency and high-specificity gene expression in brainECs through intracranial injection at various developmental stages and through intravenous administration at all postnatal stages in mice. Furthermore, we used rAAV-miniBEND to model sporadic cerebral cavernous malformations mediated byMAP3K3I441Mand AVMs mediated byBrafV600E. Somatic expression ofBrafV600Ein brainECs induced an AVM phenotype, revealing that brainEC proliferation is important for AVM development. Thus, our rAAV-miniBEND system provides a widely applicable tool for cerebrovascular disease modelling and local or global brainEC gene delivery.

Blood–brain barrier Experimental models of disease Gene delivery Transcriptional regulatory elements


N
Nature Biomedical Engineering · Oct 28, 2025

Computational design of synthetic receptors with programmable signalling activity for enhanced cancer T cell therapy

The tumour microenvironment (TME) plays a key role in tumour progression, and soluble and cellular TME components can limit CAR-T cell function and persistence. Targeting soluble TME factors to enhance anti-tumour responses of engineered T cells through chimeric receptors is not broadly explored owing to the unpredictable signalling characteristics of synthetic protein receptors. Here we develop a computational protein design platform for the de novo bottom-up assembly of allosteric receptors with programmable input–output behaviours that respond to soluble TME factors with co-stimulation and cytokine signals in T cells, called TME-sensing switch receptor for enhanced response to tumours (T-SenSER). We develop two sets of T-SenSERs targeting vascular endothelial growth factor (VEGF) or colony-stimulating factor 1 (CSF1) that are both selectively enriched in a variety of tumours. Combination of CAR and T-SenSER in human T cells enhances anti-tumour responses in models of lung cancer and multiple myeloma, in a VEGF- or CSF1-dependent manner. Our study sets the stage for the accelerated development of synthetic biosensors with custom-built sensing and responses for basic and translational cell engineering applications. A computational method designs receptors called T-SenSERs, which have predictable signalling responses to tumour microenvironment soluble factors and can be co-expressed with conventional CARs in T cells to enhance their therapeutic activity.

Cancer immunotherapy Protein design

N
Nature Biomedical Engineering · Oct 24, 2025

Implanted microelectrode arrays in reinnervated muscles allow separation of neural drives from transferred polyfunctional nerves

Targeted muscle reinnervation surgery reroutes residual nerve signals into spare muscles, enabling the recovery of neural information through electromyography (EMG). However, EMG signals are often overlapping, making the interpretation of limb functions complicated. Regenerative peripheral nerve interfaces surgically partition the nerve into individual fascicles that reinnervate specific muscle grafts, isolating distinct neural sources for precise control and interpretation of EMG signals. Here we combine targeted muscle reinnervation surgery of polyvalent nerves with a high-density microelectrode array implanted at a single site within a reinnervated muscle, and via mathematical source separation methods, we separate all neural signals that are redirected into a single muscle. In participants with upper-limb amputation, the deconvolution of EMG signals from four reinnervated muscles into motor unit spike trains revealed distinct clusters of motor neurons associated with diverse functional tasks. Our method enabled the extraction of multiple neural commands within a single reinnervated muscle, eliminating the need for surgical nerve division. This approach holds promises for enhancing control over prosthetic limbs and for understanding how the central nervous system encodes movement after reinnervation.

Biomedical engineering Computational neuroscience Machine learning Microarrays Motor neuron

N
Nature Biomedical Engineering · Oct 24, 2025

Engineered thoracic spinal cord organoids for transplantation after spinal cord injury

Stem-cell-based neural tissue engineering and spinal cord organoids show promises for spinal cord injury repair. However, the native spinal cord presents cell heterogeneity and a stereotypical spatial structure that makes difficult their recapitulation within an organoid architecture, which requires an assembly encompassing cellular composition, segmental organization and dorsoventral features. Here we engineer a thoracic vertebral segment-specific spinal cord organoid (enTsOrg) model that can precisely match the transplantation site, establish synaptic connections and enhance in vivo neuroelectric conduction. The organoids are generated from fibroblasts-derived induced pluripotent stem cells and a layered double-hydroxide matrix in a basement membrane hydrogel (Matrigel). Grafted in a spinal cord injury mouse model, enTsOrg presents advanced maturation, functionalization and organized distribution of critical neuronal subtypes with thoracic segmental heterogeneity, including various motor neuron and interneuron subtypes, that serve essentially to restore motor functions. Transplantation of enTsOrg can restructure neural circuits in paralysed animals and restore hind-limb motor function. The robust neurological function and therapeutic efficacy of enTsOrg highlight a potential avenue for organoid designing for specific anatomical regions in neurological injury treatments.

Biomaterials Spinal cord injury Stem cells


N
Nature Biomedical Engineering · Oct 23, 2025

Collagen-binding IL-12-armoured STEAP1 CAR-T cells reduce toxicity and treat prostate cancer in mouse models

Immunosuppressive microenvironments, the lack of immune infiltration, and antigen heterogeneity pose challenges for chimaeric antigen receptor (CAR)-T cell therapies applied to solid tumours. Previously, CAR-T cells were armoured with immunostimulatory molecules, such as interleukin 12 (IL-12), to overcome this issue, but faced high toxicity. Here we show that collagen-binding domain-fused IL-12 (CBD-IL-12) secreted from CAR-T cells to target human six transmembrane epithelial antigen of prostate 1 (STEAP1) is retained within murine prostate tumours. This leads to high intratumoural interferon-γ levels, without hepatotoxicity and infiltration of T cells into non-target organs compared with unmodified IL-12. Both innate and adaptive immune compartments are activated and recognize diverse tumour antigens after CBD-IL-12-armoured CAR-T cell treatment. A combination of CBD-IL-12-armoured CAR-T cells and immune checkpoint inhibitors eradicated large tumours in an established prostate cancer mouse model. In addition, human CBD-IL-12-armoured CAR-T cells showed potent anti-tumour efficacy in a 22Rv1 xenograft while reducing circulating IL-12 levels compared with unmodified IL-12-armoured CAR-T cells. CBD fusion to potent payloads for CAR-T therapy may remove obstacles to their clinical translation towards elimination of solid tumours.

Cancer immunotherapy Gene therapy Molecular medicine Protein delivery Synthetic biology

N
Nature Biomedical Engineering · Oct 22, 2025

Targeting immunosuppressive myeloid cells via implant-mediated slow release of small molecules to prevent glioblastoma recurrence

Glioblastoma is a highly aggressive brain tumour with a high risk of recurrence after surgery, even when combined with chemotherapy and radiotherapy. A major barrier to lasting treatment is the tumour’s immunosuppressive environment, which is largely dominated by myeloid cells. Here we describe the development of a biodegradable implant to sustainably release immune-modulator small molecules to reprogram tumour-infiltrating myeloid cells toward a pro-inflammatory, antitumour phenotype in the surgical cavity after tumour removal. In immunocompetent mouse models, this therapy induces interleukin-12 expression in myeloid cells without systemic cytokine elevation, and increases the infiltration of CD8+and CD4+T cells. Over 50% of mice treated (in combination with radiotherapy and chemotherapy) remain tumour-free during the experimental course (80 days). We further treated human glioblastoma explants ex vivo with the therapy and observed increased interleukin-12 expression in tumour-infiltrating myeloid cells, supporting the translational potential of this strategy. This implantable system offers a promising approach to prevent glioblastoma recurrence by activating innate immunity and sustaining immune surveillance post-surgery.

Cancer immunotherapy Cancer microenvironment CNS cancer Drug delivery Immunotherapy


N
Nature Biomedical Engineering · Oct 22, 2025

A HO-1 gene knockout using a NanoCRISPR scaffold suppresses metastasis in mouse models

Photodynamic therapy-induced immunogenic cell death has the potential to generate autologous cancer vaccines. However, the innate or evolved genetic tolerance of tumours limits the efficacy of this approach. Here we report the development of a heritable nanoplatform based on gene editing of haem oxygenase-1 (HO-1) using a NanoCRISPR/HO-1 scaffold. This platform effectively eliminates genetic tolerance to reactive oxygen species in tumours without causing adverse effects on main immune cells, resulting in a robust and durable immune response to autologous vaccine. This NanoCRISPR scaffold can inherit susceptibility to tumour progeny, transforming heterogeneous malignancies into a reactive oxyen species-sensitive phenotype. Moreover, the arginine-grafted polyethyleneimine module and CpG motif within the NanoCRISPR scaffold enhance the cancer-immune cycle by amplifying antigen generation, promoting T cell proliferation and activating adaptive immune response in cancer models. When combined with an αPD-L1 antibody, the NanoCRISPR scaffold-based heritable nanoplatform elicits antitumour immunity and durable immunological memory in vivo melanoma mouse models. This combinational therapy evokes a strong immune memory against tumour rechallenge, providing insights into the rational development of a cancer vaccine regimen.

Genetic engineering Nanoparticles








N
Nature Biomedical Engineering · Oct 03, 2025

Enzyme-converted O kidneys allow ABO-incompatible transplantation without hyperacute rejection in a human decedent model

ABO-incompatible kidney transplantation is widely used to meet the escalating need for organs. Current recipient-centric desensitization protocols involving antibody depletion through plasmapheresis increase the risk of infections, perioperative bleeding events and costs. Here we present a donor-centric desensitization protocol, converting type-A kidneys into enzyme-converted O kidneys during hypothermic perfusion to remove the A antigen from the kidneys. An ex vivo model resulted in no antibody-mediated injury. Encouraged by this, an enzyme-converted O kidney was transplanted into a type-O brain-dead recipient with a high titre of anti-A antibody, and no hyperacute rejection was observed. The graft was well tolerated with no evidence of antibody-mediated rejection for 2 days. Antibody-mediated lesions and complement deposition were found starting 3 days post-transplant, coinciding with A-antigen regeneration, and later higher Banff scores, suggesting an immune-mediated response. Single-cell sequencing confirms the elevated expression of accommodation-related genes, suggesting the potential for longer-term tolerance. This study provides a donor-centric organ engineering strategy and has the potential to broaden the reach of ABO-incompatible kidney transplantation, improving the fairness of and access to organ allocation.

Biomedical materials Kidney

N
Nature Biomedical Engineering · Oct 02, 2025

Minimally invasive implantation of scalable high-density cortical microelectrode arrays for multimodal neural decoding and stimulation

High-bandwidth brain–computer interfaces rely on invasive surgical procedures or brain-penetrating electrodes. Here we describe a cortical 1,024-channel thin-film microelectrode array and we demonstrate its minimally invasive surgical delivery that avoids craniotomy in porcine models and cadavers. We show recording and stimulation from the same electrodes to large portions of the cortical surface, and the reversibility of delivering the implants to multiple functional regions of the brain without damaging the cortical surface. We evaluate the performance of the interface for high-density neural recording and visualizing cortical surface activity at spatial and temporal resolutions and total spatial extents. We demonstrate accurate neural decoding of somatosensory, visual and volitional walking activity, and achieve focal neuromodulation through cortical stimulation at sub-millimetre scales. We report the feasibility of intraoperative use of the device in a five-patient pilot clinical study with anaesthetized and awake neurosurgical patients, characterizing the spatial scales at which sensorimotor activity and speech are represented at the cortical surface. The presented neural interface demonstrates the highly scalable nature of micro-electrocorticography and its utility for next-generation brain–computer interfaces.

Neurology Neuroscience


N
Nature Biomedical Engineering · Oct 01, 2025

Solid tumour CAR-T cells engineered with fusion proteins targeting PD-L1 for localized IL-12 delivery

Chimeric antigen receptor (CAR)-T cell efficacy in solid tumours is limited due in part to the immunosuppressive tumour microenvironment (TME). To improve antitumour responses, we hypothesized that enabling CAR-T cells to secrete bifunctional fusion proteins consisting of a cytokine modifier such as TGFβtrap, IL-15 or IL-12, combined with an immune checkpoint inhibitor such as αPD-L1, would provide tumour-localized immunomodulation to improve CAR-T cell functionality. Here we engineer CAR-T cells to secrete TGFβtrap, IL-15 or IL-12 molecules fused to αPD-L1 scFv and assess in vitro functionality and in vivo safety and efficacy in prostate and ovarian cancer models. CAR-T cells engineered with αPD-L1–IL-12 are superior in safety and efficacy compared with CAR-T cells alone and those engineered with αPD-L1 fused with TGFβtrap or IL-15. Further, αPD-L1–IL-12 engineered CAR-T cells improve T cell trafficking and tumour infiltration, and localize IFNγ production, TME modulation and antitumour responses, with reduced systemic inflammation-associated toxicities. We believe our αPD-L1–IL-12 engineering strategy presents an opportunity to improve CAR-T cell clinical efficacy and safety across multiple solid tumour types. CAR-T cells engineered with αPD-L1–IL-12 fusion proteins show antitumour activity in mouse models of prostate and ovarian cancer.

Interleukins Tumour immunology


N
Nature Biomedical Engineering · Oct 01, 2025

Large-scale visualization of α-synuclein oligomers in Parkinson’s disease brain tissue

Parkinson’s disease (PD) is a neurodegenerative condition characterized by the presence of intraneuronal aggregates containing fibrillar ɑ-synuclein known as Lewy bodies. These large end-stage species are formed by smaller soluble protein nanoscale assemblies, often termed oligomers, which are proposed as early drivers of pathogenesis. Until now, this hypothesis has remained controversial, at least in part because it has not been possible to directly visualize nanoscale assemblies in human brain tissue. Here we present Advanced Sensing of Aggregates—Parkinson’s Disease, an imaging method to generate large-scale α-synuclein aggregate maps in post-mortem human brain tissue. We combined autofluorescence suppression with single-molecule fluorescence microscopy, which together enable the detection of nanoscale α-synuclein aggregates. To demonstrate the use of this platform, we analysed ~1.2 million nanoscale aggregates from the anterior cingulate cortex in human post-mortem brain samples from patients with PD and healthy controls. Our data reveal a disease-specific shift in a subpopulation of nanoscale assemblies that represent an early feature of the proteinopathy that underlies PD. We anticipate that quantitative information about this distribution provided by Advanced Sensing of Aggregates—Parkinson’s Disease will enable mechanistic studies to reveal the pathological processes caused by α-synuclein aggregation.

High-throughput screening Nanoscale biophysics

N
Nature Biomedical Engineering · Oct 01, 2025

Targeted clearance of extracellular Tau using aptamer-armed monocytes alleviates neuroinflammation in mice with Alzheimer’s disease

Extracellular Tau determines the progression of Alzheimer’s disease, yet therapeutic strategies targeting it are hindered by poor brain delivery and limited clearance. Here we developed a Tau-clearing cell therapy based on monocytes functionalized with a high-affinity Tau-specific aptamer. The aptamer was covalently conjugated to the surface of monocytes (derived from bone marrow leucocytes and cultured under monocyte-inducing conditions) via bioorthogonal chemistry without affecting their viability or function. Upon intravenous administration in mice expressing mutant and disease-relevant human Tau, the engineered monocytes actively crossed the blood–brain barrier and accumulated in Tau-rich brain regions such as the hippocampus and striatum. They efficiently phagocytosed extracellular Tau, leading to a significant reduction in Tau burden. As a result, glial activation was suppressed, neuroinflammation was alleviated, and neuronal and mitochondrial integrity was preserved. Long-term treatment improved memory and spatial learning, without inducing toxicity or behavioural side effects. These results demonstrate that aptamer-guided monocytes can achieve targeted delivery, effective clearance and sustained neuroprotection, offering a promising strategy for therapeutic intervention in Alzheimer’s disease.

Alzheimer's disease Drug delivery


N
Nature Biomedical Engineering · Sep 30, 2025

Brain–heart–eye axis revealed by multi-organ imaging genetics and proteomics

Multi-organ research investigates interconnections among multiple human organ systems, enhancing our understanding of human aging and disease mechanisms. Here we use multi-organ imaging, individual- and summary-level genetics, and proteomics data consolidated via the MULTI Consortium to delineate a brain–heart–eye axis using brain patterns of structural covariance (PSCs), heart imaging-derived phenotypes (IDPs) and eye IDPs. We find that proteome-wide associations of the PSCs and IDPs show within-organ specificity and cross-organ interconnections. Pleiotropic effects of common single-nucleotide polymorphisms are observed across multiple organs, and key genetic parameters are estimated for single-nucleotide polymorphism-based heritability, polygenicity and selection signatures across the three organs. A gene–drug–disease network shows the potential of drug repurposing for cross-organ diseases. Co-localization and causal analyses reveal cross-organ causal relationships between PSC/IDP and chronic diseases, such as Alzheimer’s disease, heart failure and glaucoma. Finally, integrating multi-organ/omics features improves prediction for systemic disease categories and cognition compared with single-organ/omics features, providing future avenues for modelling human aging and disease.

Genetics research Heritable quantitative trait Machine learning


N
Nature Biomedical Engineering · Sep 24, 2025

Invasive neurophysiology and whole brain connectomics for neural decoding in patients with brain implants

Brain–computer interface research can inspire closed-loop neuromodulation therapies, promising spatiotemporal precision for the treatment of brain disorders. Decoding dynamic patient states from brain signals with machine learning is required to leverage this precision, but a standardized framework for invasive brain signal decoding from neural implants does not exist. Here we develop a platform that integrates brain signal decoding with magnetic resonance imaging connectomics and demonstrate its use across 123 h of invasively recorded brain data from 73 neurosurgical patients treated with brain implants for movement disorders, depression and epilepsy. We introduce connectomics-informed movement decoders that generalize across cohorts with Parkinson’s disease and epilepsy from the United States, Europe and China. We reveal network targets for emotion decoding in left prefrontal and cingulate circuits in deep brain stimulation patients with major depression. Finally, we showcase opportunities to improve seizure detection in responsive neurostimulation for epilepsy. Our study highlights the clinical use of brain signal decoding for deep brain stimulation and provides methods that allow for rapid, high-accuracy decoding for precision medicine approaches that can dynamically adapt neurotherapies in response to the individual needs of patients.

Biomedical engineering Computational neuroscience


N
Nature Biomedical Engineering · Sep 23, 2025

An immune-competent lung-on-a-chip for modelling the human severe influenza infection response

Severe influenza affects 3–5 million people worldwide each year, resulting in more than 300,000 deaths annually. However, standard-of-care antiviral therapeutics have limited effectiveness in these patients. Current preclinical models of severe influenza fail to accurately recapitulate the human immune response to severe viral infection. Here we develop an immune-competent, microvascularized, human lung-on-a-chip device to model the small airways, successfully demonstrating the cytokine storm, immune cell activation, epithelial cell damage, and other cellular- and tissue-level human immune responses to severe H1N1 infection. We find that interleukin-1β and tumour necrosis factor-α play opposing roles in the initiation and regulation of the cytokine storm associated with severe influenza. Furthermore, we discover the critical stromal–immune CXCL12–CXCR4 interaction and its role in immune response to infection. Our results underscore the importance of stromal cells and immune cells in microphysiological models of severe lung disease, describing a scalable model for severe influenza research. We expect the immune-competent human lung-on-a-chip device to enable critical discoveries in respiratory host–pathogen interactions, therapeutic side effects, vaccine potency evaluation, and crosstalk between systemic and mucosal immunity in human lung.

Biomedical engineering Influenza virus Tissue engineering


N
Nature Biomedical Engineering · Sep 22, 2025

Intranasal vaccine combining adenovirus and trimeric subunit protein provides superior immunity against SARS-CoV-2 Omicron variant

Mucosal immunity provides efficient protection against upper-airway infections, limiting viral shedding and transmission. However, currently, no nasal spray COVID-19 vaccines are approved by WHO for global use. Here we develop a two-component intranasal vaccine that combines an adenovirus vector expressing the spike protein of the XBB.1.5 variant (Ad5XBB.1.5) with a self-assembled trimeric recombinant protein derived from the receptor binding domain (RBDXBB.1.5-HR). This two-component vaccine elicits superior humoral and cellular immunity against XBB.1.5 variants compared with the individual components. It also provides protective immunity against live XBB.1.16 virus challenges in mice, and prevents XBB.1.5 virus transmission in a hamster model. Notably, the activation of the STING signalling pathway in mucosal dendritic cells is essential for the adjuvant effect of the adenovirus vector. We also incorporate another trimeric protein from the BA.5 variant (RBDBA.5-HR), creating a three-component vaccine (Ad5XBB.1.5+ RBDXBB.1.5-HR + RBDBA.5-HR) that shows enhanced broad-spectrum neutralization. The two-component vaccine demonstrates high tolerability and safety in humans, inducing enhanced mucosal immunity and high levels of neutralizing antibodies in all participants. Our findings underscore this strategy for clinical COVID-19 intranasal vaccine development.

SARS-CoV-2 Vaccines


N
Nature Biomedical Engineering · Sep 18, 2025

Systematic functional screening of switchable aptamer beacon probes

Immunoassays using affinity binders such as antibodies and aptamers are crucial for molecular biology. However, the advancement of analytical methods based on these affinity probes is often hampered by complex operational steps that can introduce errors, particularly in intricate environments such as intracellular settings and microfluidic systems. There is growing interest in developing molecular probes for wash-free assays that activate signals upon target detection. Here we report a systematic functional screening platform for switchable aptamer beacon probes that can achieve target-responsive detection. A stem–loop, hairpin-shaped beacon library was constructed on microbeads and screened using target-responsive fluorescence-activated sorting. The selected aptamer beacons exhibit strong affinities, triggering fluorescence only upon binding, thus enabling wash-free immunoassays for the detection of intracellular and membrane proteins. Computational modelling offers insights into aptamer binding and structural switching mechanisms, revealing how specific protein–aptamer interactions drive stem–loop unwinding and postbinding conformational changes critical for functional activation. This approach establishes a standardized platform for generating switchable aptameric tools, supporting their potential in advanced diagnostics and research.

Biochemical assays Biomedical engineering

N
Nature Biomedical Engineering · Sep 16, 2025

Leveraging large language and vision models for knowledge extraction from large-scale image–text colonoscopy records

The development of artificial intelligence systems for colonoscopy analysis often necessitates expert-annotated image datasets. However, limitations in dataset size and diversity impede model performance and generalization. Image–text colonoscopy records from routine clinical practice, comprising millions of images and text reports, serve as a valuable data source, although annotating them is labour intensive. Here we leverage recent advancements in large language and vision models and propose EndoKED, a data mining paradigm for deep knowledge extraction and distillation. EndoKED automates the transformation of raw colonoscopy records into image datasets with pixel-level annotation. We apply EndoKED to multicentre datasets of raw colonoscopy records (~1 million images), showing its superior performance in detecting polyps at the report and image levels, as well as annotating polyps at the pixel level. The state-of-the-art performance and generalization ability of polyp segmentation models are achieved through EndoKED pretraining. Furthermore, the EndoKED vision backbone enables data-efficient learning for optical biopsy, achieving expert-level performance in internal, external and prospective validation datasets. EndoKED is a knowledge extraction and distillation method that connects large language models and large vision models to automate the transformation of raw colonoscopy records into annotated image datasets.

Colonoscopy Computational science




N
Nature Biomedical Engineering · Sep 05, 2025

A generalist foundation model and database for open-world medical image segmentation

Vision foundation models have demonstrated vast potential in achieving generalist medical segmentation capability, providing a versatile, task-agnostic solution through a single model. However, current generalist models involve simple pre-training on various medical data containing irrelevant information, often resulting in the negative transfer phenomenon and degenerated performance. Furthermore, the practical applicability of foundation models across diverse open-world scenarios, especially in out-of-distribution (OOD) settings, has not been extensively evaluated. Here we construct a publicly accessible database, MedSegDB, based on a tree-structured hierarchy and annotated from 129 public medical segmentation repositories and 5 in-house datasets. We further propose a Generalist Medical Segmentation model (MedSegX), a vision foundation model trained with a model-agnostic Contextual Mixture of Adapter Experts (ConMoAE) for open-world segmentation. We conduct a comprehensive evaluation of MedSegX across a range of medical segmentation tasks. Experimental results indicate that MedSegX achieves state-of-the-art performance across various modalities and organ systems in in-distribution (ID) settings. In OOD and real-world clinical settings, MedSegX consistently maintains its performance in both zero-shot and data-efficient generalization, outperforming other foundation models.

Imaging Machine learning