Jun Zeng,Ming Ma,Ze Tao,Zhengsheng Rao,Chaomeng Wu,Saifu Yin,Xiaojuan Jiang,Guo Chen,Zhiling Wang,Dan Huang,Mengli Zhu,Ling Liu,Wenqian Huo,Hao Yang,Hui Guo,Gang Chen,Feng Li,Chengyu Zheng,Dajiu Huang,Peter Rahfeld,Jayachandran N Kizhakkedathu,Stephen G Withers,Xiaofeng Lu,Keqin Zhang,Tao Lin,Turun Song
{"title":"Enzyme-converted O kidneys allow ABO-incompatible transplantation without hyperacute rejection in a human decedent model.","authors":"Jun Zeng,Ming Ma,Ze Tao,Zhengsheng Rao,Chaomeng Wu,Saifu Yin,Xiaojuan Jiang,Guo Chen,Zhiling Wang,Dan Huang,Mengli Zhu,Ling Liu,Wenqian Huo,Hao Yang,Hui Guo,Gang Chen,Feng Li,Chengyu Zheng,Dajiu Huang,Peter Rahfeld,Jayachandran N Kizhakkedathu,Stephen G Withers,Xiaofeng Lu,Keqin Zhang,Tao Lin,Turun Song","doi":"10.1038/s41551-025-01513-6","DOIUrl":"https://doi.org/10.1038/s41551-025-01513-6","url":null,"abstract":"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.","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":"93 1","pages":""},"PeriodicalIF":28.1,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145215846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mark Hettick,Elton Ho,Adam J Poole,Manuel Monge,Demetrios Papageorgiou,Kazutaka Takahashi,Morgan LaMarca,Daniel Trietsch,Kyle Reed,Mark Murphy,Stephanie Rider,Kate R Gelman,Yoon Woo Byun,Joshua S Miller,Timothy Hanson,Vanessa Tolosa,Sang-Ho Lee,Sanjay Bhatia,Peter E Konrad,Michael Mager,Craig H Mermel,Benjamin I Rapoport
{"title":"Minimally invasive implantation of scalable high-density cortical microelectrode arrays for multimodal neural decoding and stimulation.","authors":"Mark Hettick,Elton Ho,Adam J Poole,Manuel Monge,Demetrios Papageorgiou,Kazutaka Takahashi,Morgan LaMarca,Daniel Trietsch,Kyle Reed,Mark Murphy,Stephanie Rider,Kate R Gelman,Yoon Woo Byun,Joshua S Miller,Timothy Hanson,Vanessa Tolosa,Sang-Ho Lee,Sanjay Bhatia,Peter E Konrad,Michael Mager,Craig H Mermel,Benjamin I Rapoport","doi":"10.1038/s41551-025-01501-w","DOIUrl":"https://doi.org/10.1038/s41551-025-01501-w","url":null,"abstract":"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.","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":"102 1","pages":""},"PeriodicalIF":28.1,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145209117","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Junwon Kang,Hamin Kim,Haewook Jang,Hyelyn Joo,Soobin Lee,Gi Yoon Lee,Hyoung Jin Kang,Youngmin Kim,Tae Hyun Kim,Sunghoon Kwon,Eun Ju Lee
{"title":"One-day rapid sterility test for human-derived biopharmaceuticals.","authors":"Junwon Kang,Hamin Kim,Haewook Jang,Hyelyn Joo,Soobin Lee,Gi Yoon Lee,Hyoung Jin Kang,Youngmin Kim,Tae Hyun Kim,Sunghoon Kwon,Eun Ju Lee","doi":"10.1038/s41551-025-01524-3","DOIUrl":"https://doi.org/10.1038/s41551-025-01524-3","url":null,"abstract":"Biopharmaceuticals are emerging as viable alternatives to chemically synthesized drugs for potential treatment to various diseases. However, many of these human-derived components cannot withstand terminal sterilization procedures, and the duration of conventional sterility testing methods often exceeds their limited shelf life. Consequently, biopharmaceuticals are now frequently administered to patients before sterility confirmation. Here we present a nanoparticle-based enrichment and rapid sterility test that can determine product sterility within a single day, mitigating clinical risks of biopharmaceuticals and maintaining therapeutic efficacies during delivery. The assay incorporates synthetic beta-2-glycoprotein I peptides for selective isolation and purification of a broad spectrum of microorganisms and a microfluidic chip designed to automatically monitor their metabolic activities via fluorescence imaging, which are inferred from the reduction of a non-toxic dye as they grow. Compared with conventional approaches, the turnaround time was substantially reduced by >58 h with 100% accuracy and a limit of detection down to a concentration of 1 colony forming unit per millilitre. We validate our approach using various forms of clinical-grade biopharmaceutical products.","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":"286 1","pages":""},"PeriodicalIF":28.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiangzhe Kong,Rui Jiao,Haowei Lin,Ruihan Guo,Wenbing Huang,Wei-Ying Ma,Zihua Wang,Yang Liu,Jianzhu Ma
{"title":"Peptide design through binding interface mimicry with PepMimic.","authors":"Xiangzhe Kong,Rui Jiao,Haowei Lin,Ruihan Guo,Wenbing Huang,Wei-Ying Ma,Zihua Wang,Yang Liu,Jianzhu Ma","doi":"10.1038/s41551-025-01507-4","DOIUrl":"https://doi.org/10.1038/s41551-025-01507-4","url":null,"abstract":"Peptides offer advantages for targeted therapy, including oral bioavailability, cellular permeability and high specificity, setting them apart from conventional small molecules and biologics. Here we develop an artificial intelligence algorithm, PepMimic, to transform a known receptor or an existing antibody of a target into a short peptide binder by mimicking the binding interfaces between targets and known binders. We apply PepMimic to drug targets PD-L1, CD38, BCMA, HER2 and CD4. Surface plasmon resonance imaging results show that 8% of the peptides exhibit dissociation constant (KD) values at the 10-8 M level, and 26 peptides achieving KD values as low as 10-9 M, substantially higher than random library screening conducted under identical conditions. We apply PepMimic to target proteins lacking available binders by first using existing algorithms to design protein binders, followed by designing peptide through simulating these artificial interfaces. We extensively validate the top-ranked peptides using tail vein injections in breast, myeloma and lung tumour mouse models. Experimental results demonstrate effective membrane binding and highlight their strong potential for clinical diagnostic imaging and targeted therapeutic applications.","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":"34 1","pages":""},"PeriodicalIF":28.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeted clearance of extracellular Tau using aptamer-armed monocytes alleviates neuroinflammation in mice with Alzheimer's disease.","authors":"Yuting Zhuo,Yao Lu,Yan Zhu,Nachuan Wen,Guangjing Zou,Hongkun Lu,Xinyu Pei,Yutong Zhang,Qiang Zhang,Xin Wang,Wenjuan Zhang,Qingyang Zhang,Zhimin Wang,Sitao Xie,Chang-Qi Li,Weihong Tan,Liping Qiu","doi":"10.1038/s41551-025-01525-2","DOIUrl":"https://doi.org/10.1038/s41551-025-01525-2","url":null,"abstract":"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.","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":"76 1","pages":""},"PeriodicalIF":28.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145203455","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
John P Murad, Lea Christian, Reginaldo Rosa, Yuwei Ren, Alyssa J Buckley, Eric Hee Jun Lee, Lupita S Lopez, Anthony K Park, Jason Yang, Yukiko Yamaguchi, Candi Trac, Lauren N Adkins, Wen-Chung Chang, Catalina Martinez, Carl H June, Stephen J Forman, Jun Ishihara, John K Lee, Lawrence A Stern, Saul J Priceman
{"title":"Solid tumour CAR-T cells engineered with fusion proteins targeting PD-L1 for localized IL-12 delivery.","authors":"John P Murad, Lea Christian, Reginaldo Rosa, Yuwei Ren, Alyssa J Buckley, Eric Hee Jun Lee, Lupita S Lopez, Anthony K Park, Jason Yang, Yukiko Yamaguchi, Candi Trac, Lauren N Adkins, Wen-Chung Chang, Catalina Martinez, Carl H June, Stephen J Forman, Jun Ishihara, John K Lee, Lawrence A Stern, Saul J Priceman","doi":"10.1038/s41551-025-01509-2","DOIUrl":"https://doi.org/10.1038/s41551-025-01509-2","url":null,"abstract":"<p><p>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 TGFβ<sup>trap</sup>, 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 TGFβ<sup>trap</sup>, 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 TGFβ<sup>trap</sup> 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.</p>","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":" ","pages":""},"PeriodicalIF":26.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206972","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Peter Neidlinger, Omar S M El Nahhas, Hannah Sophie Muti, Tim Lenz, Michael Hoffmeister, Hermann Brenner, Marko van Treeck, Rupert Langer, Bastian Dislich, Hans Michael Behrens, Christoph Röcken, Sebastian Foersch, Daniel Truhn, Antonio Marra, Oliver Lester Saldanha, Jakob Nikolas Kather
{"title":"Benchmarking foundation models as feature extractors for weakly supervised computational pathology.","authors":"Peter Neidlinger, Omar S M El Nahhas, Hannah Sophie Muti, Tim Lenz, Michael Hoffmeister, Hermann Brenner, Marko van Treeck, Rupert Langer, Bastian Dislich, Hans Michael Behrens, Christoph Röcken, Sebastian Foersch, Daniel Truhn, Antonio Marra, Oliver Lester Saldanha, Jakob Nikolas Kather","doi":"10.1038/s41551-025-01516-3","DOIUrl":"https://doi.org/10.1038/s41551-025-01516-3","url":null,"abstract":"<p><p>Numerous pathology foundation models have been developed to extract clinically relevant information. There is currently limited literature independently evaluating these foundation models on external cohorts and clinically relevant tasks to uncover adjustments for future improvements. Here we benchmark 19 histopathology foundation models on 13 patient cohorts with 6,818 patients and 9,528 slides from lung, colorectal, gastric and breast cancers. The models were evaluated on weakly supervised tasks related to biomarkers, morphological properties and prognostic outcomes. We show that a vision-language foundation model, CONCH, yielded the highest overall performance when compared with vision-only foundation models, with Virchow2 as close second, although its superior performance was less pronounced in low-data scenarios and low-prevalence tasks. The experiments reveal that foundation models trained on distinct cohorts learn complementary features to predict the same label, and can be fused to outperform the current state of the art. An ensemble combining CONCH and Virchow2 predictions outperformed individual models in 55% of tasks, leveraging their complementary strengths in classification scenarios. Moreover, our findings suggest that data diversity outweighs data volume for foundation models.</p>","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":" ","pages":""},"PeriodicalIF":26.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rebecca Andrews, Bin Fu, Christina E Toomey, Jonathan C Breiter, Joanne Lachica, Joseph S Beckwith, Ru Tian, Emma E Brock, Lisa-Maria Needham, Gregory J Chant, Camille Loiseau, Angèle Deconfin, Kenza Baspin, Rebeka Popovic, James Evans, Yen Goh, Begüm Kurt, Lenart Senicar, Marisa Edmonds, Tim Bartels, Nora Bengoa-Vergniory, Peter J Magill, Zane Jaunmuktane, Oliver J Freeman, Benjamin J M Taylor, John Hardy, Tammaryn Lashley, Mina Ryten, Michele Vendruscolo, Nicholas W Wood, Lucien E Weiss, Sonia Gandhi, Steven F Lee
{"title":"Large-scale visualization of α-synuclein oligomers in Parkinson's disease brain tissue.","authors":"Rebecca Andrews, Bin Fu, Christina E Toomey, Jonathan C Breiter, Joanne Lachica, Joseph S Beckwith, Ru Tian, Emma E Brock, Lisa-Maria Needham, Gregory J Chant, Camille Loiseau, Angèle Deconfin, Kenza Baspin, Rebeka Popovic, James Evans, Yen Goh, Begüm Kurt, Lenart Senicar, Marisa Edmonds, Tim Bartels, Nora Bengoa-Vergniory, Peter J Magill, Zane Jaunmuktane, Oliver J Freeman, Benjamin J M Taylor, John Hardy, Tammaryn Lashley, Mina Ryten, Michele Vendruscolo, Nicholas W Wood, Lucien E Weiss, Sonia Gandhi, Steven F Lee","doi":"10.1038/s41551-025-01496-4","DOIUrl":"https://doi.org/10.1038/s41551-025-01496-4","url":null,"abstract":"<p><p>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.</p>","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":" ","pages":""},"PeriodicalIF":26.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145207012","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aleix Boquet-Pujadas, Filippos Anagnostakis, Michael R Duggan, Cassandra M Joynes, Arthur W Toga, Zhijian Yang, Keenan A Walker, Christos Davatzikos, Junhao Wen
{"title":"Brain-heart-eye axis revealed by multi-organ imaging genetics and proteomics.","authors":"Aleix Boquet-Pujadas, Filippos Anagnostakis, Michael R Duggan, Cassandra M Joynes, Arthur W Toga, Zhijian Yang, Keenan A Walker, Christos Davatzikos, Junhao Wen","doi":"10.1038/s41551-025-01506-5","DOIUrl":"https://doi.org/10.1038/s41551-025-01506-5","url":null,"abstract":"<p><p>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.</p>","PeriodicalId":19063,"journal":{"name":"Nature Biomedical Engineering","volume":" ","pages":""},"PeriodicalIF":26.8,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145200229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}