Background: Reactive astrocytes form a chemical and mechanical glial scar that inhibits neuro-regeneration after stroke. Astrocyte heterogeneity is accompanied by changes in morphology and mechanical properties altering during scar formation after injury. This work aimed to elucidate the relationship between glial scar stiffness and astrocyte subtype transformation. Methods: Astrocyte-specific archaerhodopsin-3 and channelrhodopsin-2 knock-in C57BL/6J mice underwent distal MCAO. Atomic force microscopy, ultrasound elastography and synchrotron radiation were used to determine changes in glial scar stiffness. A proteomic analysis of astrocyte subtypes was performed ex vitro using single-cell laser capture microdissection-MS. Furthermore, optogenetics was employed in vivo to reduce the glial scar stiffness, thereby facilitating neural regeneration following brain injury. Results: Glial scar stiffness systematically increases following stroke and correlates with an increased number of Wnt7b+ fibrotic astrocytes. Furthermore, these results indicate that Piezo1 is the key regulator of astrocytic stiffness and anisotropy, which contributes to the glial scar stiffness in the peri-infarct area. The downregulation of Piezo1 expression promotes activation of the Wnt7b-Ca2+ nonclassical signaling pathway to modulate cytoskeletal reorganization. Finally, the specific optogenetic inhibition of Ca2+ signaling in astrocytes can effectively reduce glial scar stiffness by decreasing the proportion of Wn7b+ astrocytes, which further promotes neuro-regeneration and improves the recovery of motor function after ischemic stroke. Conclusions: This study successfully revealed astrocyte subtype transformation as a key determinant of glial scar physical barrier formation after stroke and highlighted Piezo1 as a potential therapeutic target for modulating the mechanical microenvironment post-injury.
{"title":"Piezo1 induces Wnt7b<sup>+</sup> astrocytes transformation to modulate glial scar stiffness and neuro-regeneration after stroke.","authors":"Shengju Wu, Wenjie Hao, Qian Suo, Qijie Lu, Ze Liu, Yang Qianbo Yao, Rubing Shi, Khan Haroon, Yuewei Chen, Xinfa Shao, Qinqin Wang, Chen Li, Qun Xu, Wanlu Li, Yaohui Tang, Guo-Yuan Yang, Zhijun Zhang","doi":"10.7150/thno.120838","DOIUrl":"10.7150/thno.120838","url":null,"abstract":"<p><p><b>Background:</b> Reactive astrocytes form a chemical and mechanical glial scar that inhibits neuro-regeneration after stroke. Astrocyte heterogeneity is accompanied by changes in morphology and mechanical properties altering during scar formation after injury. This work aimed to elucidate the relationship between glial scar stiffness and astrocyte subtype transformation. <b>Methods:</b> Astrocyte-specific archaerhodopsin-3 and channelrhodopsin-2 knock-in C57BL/6J mice underwent distal MCAO. Atomic force microscopy, ultrasound elastography and synchrotron radiation were used to determine changes in glial scar stiffness. A proteomic analysis of astrocyte subtypes was performed <i>ex vitro</i> using single-cell laser capture microdissection-MS. Furthermore, optogenetics was employed <i>in vivo</i> to reduce the glial scar stiffness, thereby facilitating neural regeneration following brain injury. <b>Results:</b> Glial scar stiffness systematically increases following stroke and correlates with an increased number of Wnt7b<sup>+</sup> fibrotic astrocytes. Furthermore, these results indicate that Piezo1 is the key regulator of astrocytic stiffness and anisotropy, which contributes to the glial scar stiffness in the peri-infarct area. The downregulation of Piezo1 expression promotes activation of the Wnt7b-Ca<sup>2+</sup> nonclassical signaling pathway to modulate cytoskeletal reorganization. Finally, the specific optogenetic inhibition of Ca<sup>2+</sup> signaling in astrocytes can effectively reduce glial scar stiffness by decreasing the proportion of Wn7b<sup>+</sup> astrocytes, which further promotes neuro-regeneration and improves the recovery of motor function after ischemic stroke. <b>Conclusions:</b> This study successfully revealed astrocyte subtype transformation as a key determinant of glial scar physical barrier formation after stroke and highlighted Piezo1 as a potential therapeutic target for modulating the mechanical microenvironment post-injury.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"668-688"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674932/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678885","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment, where they facilitate tumor progression, angiogenesis, immune evasion, and treatment resistance, highlighting the urgent need for CAF-targeted strategies for high-performance tumor therapy. Recent nanomedicine approaches have shown promise in CAFs targeting in order to achieve precise targeting, spatiotemporal control of drug release, and enhanced drug penetration into dense fibrotic stroma. Accordingly, this review summarizes emerging nanotechnologies that address challenges through the development of functional nanomaterials for CAFs targeting, including polymers, metal and non-metal inorganic nanoparticles (NPs), cell membrane-based NPs, and protein-based NPs. Specifically, various therapeutic approaches such as direct CAFs depletion, signaling pathway modulation in CAFs, and CAFs reprogramming by using these nanomedicines are discussed. Furthermore, potential avenues for future studies, including the development of versatile nanosystems and the exploration of personalized treatment regimens, and challenges of advanced functional nanomaterials are involved as well. We hope that this review will offer new insights into cancer therapy and advance the development of clinically applicable CAF-targeted nanomedicines.
{"title":"Nanomedicine for targeting cancer-associated fibroblasts in cancer therapy.","authors":"Zi-Yi Chen, Han-Zhe Liu, Zheng-Jun Shang, Guo-Feng Luo, Xian-Zheng Zhang","doi":"10.7150/thno.120283","DOIUrl":"10.7150/thno.120283","url":null,"abstract":"<p><p>Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment, where they facilitate tumor progression, angiogenesis, immune evasion, and treatment resistance, highlighting the urgent need for CAF-targeted strategies for high-performance tumor therapy. Recent nanomedicine approaches have shown promise in CAFs targeting in order to achieve precise targeting, spatiotemporal control of drug release, and enhanced drug penetration into dense fibrotic stroma. Accordingly, this review summarizes emerging nanotechnologies that address challenges through the development of functional nanomaterials for CAFs targeting, including polymers, metal and non-metal inorganic nanoparticles (NPs), cell membrane-based NPs, and protein-based NPs. Specifically, various therapeutic approaches such as direct CAFs depletion, signaling pathway modulation in CAFs, and CAFs reprogramming by using these nanomedicines are discussed. Furthermore, potential avenues for future studies, including the development of versatile nanosystems and the exploration of personalized treatment regimens, and challenges of advanced functional nanomaterials are involved as well. We hope that this review will offer new insights into cancer therapy and advance the development of clinically applicable CAF-targeted nanomedicines.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 3","pages":"1545-1576"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679702/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jae Yun Kim, Han-Seop Kim, Binna Seol, Ji Eun Choi, Ji-Young Lee, Yee Sook Cho
Rationale: Natural killer (NK) cells are emerging as a promising source of immunomodulatory secretomes with regenerative potential. However, heterogeneity in primary NK cell populations limits the reproducibility of NK-derived cell-free therapies. To address this, we developed directly reprogrammed NK (drNK) cells with a stable CD56brightCD16bright phenotype and investigated the therapeutic potential of their conditioned medium (drNK-CM) in wound healing, focusing on underlying molecular mechanisms such as chemokine signaling and angiogenesis. Methods: drNK cells were generated by transcription factor-mediated reprogramming (OCT4, SOX2, KLF4, MYC) and characterized via flow cytometry and RNA-seq. The secretome profile of drNK-CM was evaluated using proteomic analysis. Human epidermal keratinocytes (HEKs), dermal fibroblasts (HDFs), and endothelial cells (HUVECs) were treated with drNK-CM to assess proliferation, migration, and extracellular matrix (ECM) remodeling. Chemokine receptor involvement was evaluated using CCR1, CCR3, and CCR5 antagonists. In vivo efficacy was tested in mouse excisional wound models, with histological and immunofluorescence evaluation of angiogenesis, re-epithelialization, and collagen deposition. Results: drNK-CM significantly promoted proliferation and migration of HEKs, HDFs, and HUVECs, accompanied by enhanced expression of Type I/III collagen, VEGF, and MMPs. Transcriptomic profiling revealed that drNKs uniquely upregulated genes associated with ECM remodeling, chemokine signaling (CCL3/4/5), and angiogenesis. Notably, CCR5 inhibition by maraviroc abrogated drNK-CM-induced cell migration and delayed wound closure in vivo, highlighting the central role of the CCL3/4/5-CCR5 axis. Furthermore, drNK-CM activated AKT and ERK pathways and promoted anti-inflammatory macrophage polarization. In vivo application of drNK-CM accelerated wound closure, improved neovascularization, and supported organized tissue regeneration compared to controls. Conclusion: This study demonstrates that drNK-CM enhances wound healing through coordinated actions on epithelial, stromal, and endothelial compartments. The reparative effects are primarily mediated via the CCL3/4/5-CCR5 signaling axis and pro-angiogenic cascades. Given their consistent phenotype and reproducible secretome, drNKs represent a scalable and safe source for cell-free regenerative therapeutics.
{"title":"Cell-free secretome of CD56<sup>bright</sup>CD16<sup>bright</sup> directly reprogrammed NK cells enhances wound healing via CCL3/4/5-CCR5 signaling.","authors":"Jae Yun Kim, Han-Seop Kim, Binna Seol, Ji Eun Choi, Ji-Young Lee, Yee Sook Cho","doi":"10.7150/thno.120219","DOIUrl":"10.7150/thno.120219","url":null,"abstract":"<p><p><b>Rationale:</b> Natural killer (NK) cells are emerging as a promising source of immunomodulatory secretomes with regenerative potential. However, heterogeneity in primary NK cell populations limits the reproducibility of NK-derived cell-free therapies. To address this, we developed directly reprogrammed NK (drNK) cells with a stable CD56<sup>bright</sup>CD16<sup>bright</sup> phenotype and investigated the therapeutic potential of their conditioned medium (drNK-CM) in wound healing, focusing on underlying molecular mechanisms such as chemokine signaling and angiogenesis. <b>Methods:</b> drNK cells were generated by transcription factor-mediated reprogramming (OCT4, SOX2, KLF4, MYC) and characterized via flow cytometry and RNA-seq. The secretome profile of drNK-CM was evaluated using proteomic analysis. Human epidermal keratinocytes (HEKs), dermal fibroblasts (HDFs), and endothelial cells (HUVECs) were treated with drNK-CM to assess proliferation, migration, and extracellular matrix (ECM) remodeling. Chemokine receptor involvement was evaluated using CCR1, CCR3, and CCR5 antagonists. <i>In vivo</i> efficacy was tested in mouse excisional wound models, with histological and immunofluorescence evaluation of angiogenesis, re-epithelialization, and collagen deposition. <b>Results:</b> drNK-CM significantly promoted proliferation and migration of HEKs, HDFs, and HUVECs, accompanied by enhanced expression of Type I/III collagen, VEGF, and MMPs. Transcriptomic profiling revealed that drNKs uniquely upregulated genes associated with ECM remodeling, chemokine signaling (CCL3/4/5), and angiogenesis. Notably, CCR5 inhibition by maraviroc abrogated drNK-CM-induced cell migration and delayed wound closure <i>in vivo</i>, highlighting the central role of the CCL3/4/5-CCR5 axis. Furthermore, drNK-CM activated AKT and ERK pathways and promoted anti-inflammatory macrophage polarization. <i>In vivo</i> application of drNK-CM accelerated wound closure, improved neovascularization, and supported organized tissue regeneration compared to controls. <b>Conclusion:</b> This study demonstrates that drNK-CM enhances wound healing through coordinated actions on epithelial, stromal, and endothelial compartments. The reparative effects are primarily mediated via the CCL3/4/5-CCR5 signaling axis and pro-angiogenic cascades. Given their consistent phenotype and reproducible secretome, drNKs represent a scalable and safe source for cell-free regenerative therapeutics.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"952-969"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12675002/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701723","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yan Xiang, Ao Du, Zhen Wang, Hongyuan Pan, Kefei Yuan
Short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, serve as pivotal metabolites within the tumor microenvironment (TME), playing essential roles in modulating tumor progression. Although the biological functions and mechanisms of SCFAs in the TME show some overlap, each SCFA also exerts some distinct regulatory effects on tumors and TME. Notably, even a single SCFA may exhibit pleiotropic effects across different cancer types or under varying conditions within the same malignancy. Consequently, according to the different metabolic microenvironment of patients, precise modulation of SCFA levels could effectively suppress tumor progression. Furthermore, SCFAs have been shown to potentiate the therapeutic efficacy of immunotherapy, radiotherapy, and chemotherapy. This review systematically outlines the sources, biological functions, and mechanisms of different SCFAs in the TME, while exploring potential therapeutic strategies based on SCFA modulation. These insights offer novel perspectives and directions for future research and clinical cancer therapy.
{"title":"Short-chain fatty acids in the tumor microenvironment: from molecular mechanisms to cancer therapy.","authors":"Yan Xiang, Ao Du, Zhen Wang, Hongyuan Pan, Kefei Yuan","doi":"10.7150/thno.119304","DOIUrl":"10.7150/thno.119304","url":null,"abstract":"<p><p>Short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, serve as pivotal metabolites within the tumor microenvironment (TME), playing essential roles in modulating tumor progression. Although the biological functions and mechanisms of SCFAs in the TME show some overlap, each SCFA also exerts some distinct regulatory effects on tumors and TME. Notably, even a single SCFA may exhibit pleiotropic effects across different cancer types or under varying conditions within the same malignancy. Consequently, according to the different metabolic microenvironment of patients, precise modulation of SCFA levels could effectively suppress tumor progression. Furthermore, SCFAs have been shown to potentiate the therapeutic efficacy of immunotherapy, radiotherapy, and chemotherapy. This review systematically outlines the sources, biological functions, and mechanisms of different SCFAs in the TME, while exploring potential therapeutic strategies based on SCFA modulation. These insights offer novel perspectives and directions for future research and clinical cancer therapy.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 3","pages":"1143-1163"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679088/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701752","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Preethi Korangath, Hayden Carlton, Toby Sanders, Olivia C Sehl, Suqi Ke, Abdul Rahman Mohtasebzadeh, Lyndsey Werhane, Cordula Grüttner, Chen Hu, Kathleen Gabrielson, Patrick W Goodwill, Robert Ivkov
Early detection of metastatic disease improves cancer survival, yet existing modalities are limited in their detection capabilities. We propose that magnetic particle imaging (MPI), an emerging technology, can be used for early detection of primary tumors and metastases. MPI detects minute quantities of magnetic particles that act as "cold tracers" which accumulate in areas of high immune activity. Methods: Pegylated Synomag® nanoparticles were intravenously injected into mouse models of breast cancer bearing primary tumors and spontaneously developed lung metastases. After 72 h, mice were subjected to three-dimensional MPI followed by structural imaging for co-registration. Non-tumor bearing mice served as controls for background signal correction and toxicity analysis. Animals were then sacrificed to collect tumors and organs of interest for two-dimensional MPI scans before fixing them for histopathological evaluation by hematoxylin and Eosin (H&E), Prussian blue, and immunohistochemistry staining. To further substantiate our findings towards clinical translation, tumor phantoms with nanoparticles were evaluated in a newly-built human scale MPI. Results: Pegylated Synomag® nanoparticles showed a strong signal in both in vitro and in vivo models. Multiple macro and micro metastatic sites were identified by MPI and later confirmed by histology. Ex vivo quantitative analysis showed MPI can detect metastasis with high specificity and sensitivity, with positive correlations between tumor burden and macrophage population in the tumor microenvironment. Towards clinical translation, we also demonstrate nanoparticle detection in tumor phantoms using a human-scale MPI. Conclusion: MPI using Pegylated Synomag® nanoparticles can successfully detect primary tumors and micrometastases away from large organs of the reticuloendothelial system. Nanoparticles were found in the tumor microenvironment, associated with stromal and immune cells, especially macrophages. This provides evidence to use MPI for noninvasive detection of highly inflammatory tumors and metastasis, as well as exploring their potential for other inflammatory diseases.
{"title":"Nanoparticle retention enables non-invasive detection of metastases by magnetic particle imaging in murine breast cancer models.","authors":"Preethi Korangath, Hayden Carlton, Toby Sanders, Olivia C Sehl, Suqi Ke, Abdul Rahman Mohtasebzadeh, Lyndsey Werhane, Cordula Grüttner, Chen Hu, Kathleen Gabrielson, Patrick W Goodwill, Robert Ivkov","doi":"10.7150/thno.122259","DOIUrl":"10.7150/thno.122259","url":null,"abstract":"<p><p>Early detection of metastatic disease improves cancer survival, yet existing modalities are limited in their detection capabilities. We propose that magnetic particle imaging (MPI), an emerging technology, can be used for early detection of primary tumors and metastases. MPI detects minute quantities of magnetic particles that act as \"cold tracers\" which accumulate in areas of high immune activity. <b>Methods:</b> Pegylated Synomag® nanoparticles were intravenously injected into mouse models of breast cancer bearing primary tumors and spontaneously developed lung metastases. After 72 h, mice were subjected to three-dimensional MPI followed by structural imaging for co-registration. Non-tumor bearing mice served as controls for background signal correction and toxicity analysis. Animals were then sacrificed to collect tumors and organs of interest for two-dimensional MPI scans before fixing them for histopathological evaluation by hematoxylin and Eosin (H&E), Prussian blue, and immunohistochemistry staining. To further substantiate our findings towards clinical translation, tumor phantoms with nanoparticles were evaluated in a newly-built human scale MPI. <b>Results:</b> Pegylated Synomag® nanoparticles showed a strong signal in both <i>in vitro</i> and <i>in vivo</i> models. Multiple macro and micro metastatic sites were identified by MPI and later confirmed by histology. <i>Ex vivo</i> quantitative analysis showed MPI can detect metastasis with high specificity and sensitivity, with positive correlations between tumor burden and macrophage population in the tumor microenvironment. Towards clinical translation, we also demonstrate nanoparticle detection in tumor phantoms using a human-scale MPI. <b>Conclusion:</b> MPI using Pegylated Synomag® nanoparticles can successfully detect primary tumors and micrometastases away from large organs of the reticuloendothelial system. Nanoparticles were found in the tumor microenvironment, associated with stromal and immune cells, especially macrophages. This provides evidence to use MPI for noninvasive detection of highly inflammatory tumors and metastasis, as well as exploring their potential for other inflammatory diseases.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 5","pages":"2136-2155"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12712802/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145805535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Siaka Fadera, Cristian Antonio Wieczorek Villas Boas, Yimei Yue, Zhaoning Gu, Jinyun Yuan, Debolina De, Buck E Rogers, Arash Nazeri, Hong Chen
Background: The intranasal (IN) route offers a promising noninvasive strategy for central nervous system drug delivery bypassing the blood-brain barrier and reducing systemic exposure. However, its clinical translation is limited by low delivery efficiency and a lack of regional specificity in the brain. Here, we present the first demonstration of focused ultrasound-mediated intranasal delivery (FUSIN) in a large-animal model to address these limitations. Methods: Pigs were used to develop and characterize IN delivery, evaluate systemic exposure in major organs, and assess the feasibility and safety of FUSIN for delivering a therapeutic antibody, anti-programmed death-ligand 1 antibody (aPD-L1). The IN delivery was performed using a catheter-based approach, and successful delivery was confirmed with gadolinium-based contrast agents in combination with magnetic resonance imaging (MRI). Systemic exposure was assessed following IN administration of fluorescently labeled IRDye 800CW-aPD-L1, and its biodistribution was compared with intravenous (IV) injection of IRDye 700CW-aPD-L1. FUSIN delivery was performed by applying focused ultrasound (FUS) to predefined brain targets following IN administration to enhance local antibody accumulation. Delivery outcomes were assessed by ex vivo fluorescence imaging, followed by immunofluorescence staining, and safety was evaluated using susceptibility-weighted imaging (SWI). Results: IN delivery targeted the olfactory epithelium region and resulted in significant accumulation of 800CW-aPD-L1 in brain regions associated with the olfactory and trigeminal pathways, while markedly reducing off-target deposition in peripheral organs compared to IV administration. The application of FUS significantly increased local antibody accumulation at the targeted sites compared to contralateral non-sonicated controls. Immunofluorescence imaging revealed FUS-enhanced transport of the antibody from perivascular spaces into the brain interstitial space. SWI detected microhemorrhages under the current FUS parameters, highlighting the need for optimization to ensure safety. Conclusion: This study demonstrates the feasibility of FUSIN for noninvasive, region-specific brain drug delivery with minimized systemic exposure in a large-animal model.
{"title":"Focused ultrasound-enhanced nose-to-brain delivery of a therapeutic antibody in a large-animal model.","authors":"Siaka Fadera, Cristian Antonio Wieczorek Villas Boas, Yimei Yue, Zhaoning Gu, Jinyun Yuan, Debolina De, Buck E Rogers, Arash Nazeri, Hong Chen","doi":"10.7150/thno.124354","DOIUrl":"10.7150/thno.124354","url":null,"abstract":"<p><p><b>Background:</b> The intranasal (IN) route offers a promising noninvasive strategy for central nervous system drug delivery bypassing the blood-brain barrier and reducing systemic exposure. However, its clinical translation is limited by low delivery efficiency and a lack of regional specificity in the brain. Here, we present the first demonstration of focused ultrasound-mediated intranasal delivery (FUSIN) in a large-animal model to address these limitations. <b>Methods:</b> Pigs were used to develop and characterize IN delivery, evaluate systemic exposure in major organs, and assess the feasibility and safety of FUSIN for delivering a therapeutic antibody, anti-programmed death-ligand 1 antibody (aPD-L1). The IN delivery was performed using a catheter-based approach, and successful delivery was confirmed with gadolinium-based contrast agents in combination with magnetic resonance imaging (MRI). Systemic exposure was assessed following IN administration of fluorescently labeled IRDye 800CW-aPD-L1, and its biodistribution was compared with intravenous (IV) injection of IRDye 700CW-aPD-L1. FUSIN delivery was performed by applying focused ultrasound (FUS) to predefined brain targets following IN administration to enhance local antibody accumulation. Delivery outcomes were assessed by <i>ex vivo</i> fluorescence imaging, followed by immunofluorescence staining, and safety was evaluated using susceptibility-weighted imaging (SWI). <b>Results:</b> IN delivery targeted the olfactory epithelium region and resulted in significant accumulation of 800CW-aPD-L1 in brain regions associated with the olfactory and trigeminal pathways, while markedly reducing off-target deposition in peripheral organs compared to IV administration. The application of FUS significantly increased local antibody accumulation at the targeted sites compared to contralateral non-sonicated controls. Immunofluorescence imaging revealed FUS-enhanced transport of the antibody from perivascular spaces into the brain interstitial space. SWI detected microhemorrhages under the current FUS parameters, highlighting the need for optimization to ensure safety. <b>Conclusion:</b> This study demonstrates the feasibility of FUSIN for noninvasive, region-specific brain drug delivery with minimized systemic exposure in a large-animal model.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 5","pages":"2156-2169"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12712798/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145805570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Radiotherapy resistance in breast cancer remains a major clinical challenge. The key molecular determinants and cellular populations driving this resistance are not fully understood. Methods: A radiotherapy resistance (RR) gene panel was identified from TCGA-BRCA and GSE120798 cohorts. Single-cell and spatial transcriptomics characterized RRhigh epithelial cells (RRhighepi). A prognostic model, named SuperPC and StepCox-based Radiotherapy Resistance model (SSRR), was built via machine learning and Mendelian randomization. Functional roles of Prolyl 4-Hydroxylase Subunit Alpha 2 (P4HA2) were validated in vitro. Results: The RR gene panel was upregulated in tumors and enriched for cell cycle pathways. RRhighepi cells exhibited elevated stemness, activated cell cycle and metabolic programs, and enhanced DNA damage repair. RRhighepi represented a developmental origin and communicated with endothelial cells. The SSRR model stratified patients into high-risk groups with poorer survival and distinct therapeutic responses. P4HA2, a key model gene, was upregulated in multiple cancers. P4HA2 knockdown suppressed proliferation, invasion, and colony formation, and synergized with radiotherapy to reduce stemness and enhance DNA damage. WGCNA confirmed co-module membership of P4HA2 and the RR panel. Conclusions: This study, through multi-omics analysis, proposes a potential mechanistic model associated with radiotherapy resistance in breast cancer. P4HA2 is a potential therapeutic target that sensitizes breast cancer to radiotherapy. The RR gene panel and SSRR model provide insights into resistance mechanisms and prognostic stratification.
{"title":"Single-cell and spatial transcriptomics reveal P4HA2-mediated radiotherapy resistance mechanisms in breast cancer.","authors":"Huimin Li, Junzhi Liu, Yuheng Jiao, Fengyu Xu, Shurui Wang, Qiang Tang","doi":"10.7150/thno.121257","DOIUrl":"10.7150/thno.121257","url":null,"abstract":"<p><p><b>Background:</b> Radiotherapy resistance in breast cancer remains a major clinical challenge. The key molecular determinants and cellular populations driving this resistance are not fully understood. <b>Methods:</b> A radiotherapy resistance (RR) gene panel was identified from TCGA-BRCA and GSE120798 cohorts. Single-cell and spatial transcriptomics characterized RRhigh epithelial cells (RRhighepi). A prognostic model, named SuperPC and StepCox-based Radiotherapy Resistance model (SSRR), was built via machine learning and Mendelian randomization. Functional roles of Prolyl 4-Hydroxylase Subunit Alpha 2 (<i>P4HA2</i>) were validated <i>in vitro</i>. <b>Results:</b> The RR gene panel was upregulated in tumors and enriched for cell cycle pathways. RRhighepi cells exhibited elevated stemness, activated cell cycle and metabolic programs, and enhanced DNA damage repair. RRhighepi represented a developmental origin and communicated with endothelial cells. The SSRR model stratified patients into high-risk groups with poorer survival and distinct therapeutic responses. <i>P4HA2</i>, a key model gene, was upregulated in multiple cancers. <i>P4HA2</i> knockdown suppressed proliferation, invasion, and colony formation, and synergized with radiotherapy to reduce stemness and enhance DNA damage. WGCNA confirmed co-module membership of <i>P4HA2</i> and the RR panel. <b>Conclusions:</b> This study, through multi-omics analysis, proposes a potential mechanistic model associated with radiotherapy resistance in breast cancer. <i>P4HA2</i> is a potential therapeutic target that sensitizes breast cancer to radiotherapy. The RR gene panel and SSRR model provide insights into resistance mechanisms and prognostic stratification.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 5","pages":"2192-2220"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12712805/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145805344","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The impaired healing of diabetic wounds is caused by complex multifactorial pathologies and conventional therapeutic approaches often show limited efficacy. In recent years, stimulus-responsive hydrogels based on cascade reactions have become a promising approach in the management of diabetic wounds. These hydrogels are designed to react to particular characteristics of the wound microenvironment, such as glucose concentration, pH, reactive oxygen species (ROS) and enzyme activity, allowing spatiotemporally controlled drug release and synergistic multi-target control. This review focuses on the recent development in understanding of the pathophysiology of diabetic wounds, immune microenvironment modulation, and the development of stimuli-responsive cascade hydrogels, as well as the challenges. By integrating responsive moieties, these hydrogels dynamically control the polarization of immune cells and scavenging of ROS. Furthermore, cascade systems, from single-step to multistep design, enable precise spatiotemporal activation and coordinate antibacterial, antioxidant and pro-regenerative effects. Additionally, emerging technologies such as AI-assisted modeling, biosensing-guided feedback, and organ-on-a-chip platforms have great potential to improve the rational design and predictive validation of cascade hydrogel systems, paving the way for intelligent and personalized diabetic wound therapies.
{"title":"Stimuli-responsive hydrogels based on cascade reactions: a novel strategy to promote the efficient repair of diabetic wounds.","authors":"Jialin Jia, Xiaosu Wang, Tao Zhang, Qingxia Sun, Shude Yang, Wenna Wu","doi":"10.7150/thno.126282","DOIUrl":"10.7150/thno.126282","url":null,"abstract":"<p><p>The impaired healing of diabetic wounds is caused by complex multifactorial pathologies and conventional therapeutic approaches often show limited efficacy. In recent years, stimulus-responsive hydrogels based on cascade reactions have become a promising approach in the management of diabetic wounds. These hydrogels are designed to react to particular characteristics of the wound microenvironment, such as glucose concentration, pH, reactive oxygen species (ROS) and enzyme activity, allowing spatiotemporally controlled drug release and synergistic multi-target control. This review focuses on the recent development in understanding of the pathophysiology of diabetic wounds, immune microenvironment modulation, and the development of stimuli-responsive cascade hydrogels, as well as the challenges. By integrating responsive moieties, these hydrogels dynamically control the polarization of immune cells and scavenging of ROS. Furthermore, cascade systems, from single-step to multistep design, enable precise spatiotemporal activation and coordinate antibacterial, antioxidant and pro-regenerative effects. Additionally, emerging technologies such as AI-assisted modeling, biosensing-guided feedback, and organ-on-a-chip platforms have great potential to improve the rational design and predictive validation of cascade hydrogel systems, paving the way for intelligent and personalized diabetic wound therapies.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"3105-3135"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775984/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Splicing factors play pivotal roles in mRNA processing and are implicated in tumor progression. The aberrant expression of splicing factors is closely associated with the invasiveness and secretion profiles of pituitary neuroendocrine tumors (PitNETs). In this study, we explored the involvement of splicing factors in PIT1-lineage PitNET progression and assessed the feasibility of targeting the splicing process as a therapeutic approach. Methods: Statistical data on PitNET subtypes were obtained from the National Brain Tumor Registry of China (NBTRC), and gene expression analysis was conducted on 40 clinical samples collected for this study. Transcriptome analysis and RNA immunoprecipitation sequencing (RIP-seq) were utilized to examine FUS-mediated alternative splicing and to identify mRNA binding sites in PitNET cells. Minigene splicing assays were employed to confirm the specific exonic and intronic regions. Additionally, Annexin V/PI assays and JC-1 staining were conducted to evaluate apoptosis. Results: The expression of the splicing factor FUS was elevated in PIT1-lineage PitNETs and was correlated with increased proliferative capacity and reduced apoptosis levels. Transcriptome sequencing revealed that the knockdown of FUS led to extensive exon skipping and activated the p53 pathway. In addition to RIP-seq analysis, these findings suggest that FUS contributes to the inclusion of exon 3 to generate full-length MDM2, a well-established negative regulator of p53. Antisense oligonucleotides (ASOs) specifically designed to target binding sequences on pre-mRNAs effectively disrupted the FUS-mediated splicing process, consequently impeding the progression of PitNETs. Conclusions: Our study elucidated the critical function of FUS as a splicing factor in PitNETs. Furthermore, we illustrated that targeting the splicing mechanism associated with MDM2 could restore p53 levels, thereby impeding the progression of PitNETs. This discovery presents a potentially novel strategy for the clinical management of PIT1-lineage PitNETs.
{"title":"Splicing factor FUS facilitates the progression of PIT1-lineage PitNETs by upregulating MDM2.","authors":"Xu Wang, Jiang Li, Chenggang Jiang, Chengkai Zhang, Linhao Yuan, Tieqiang Zhang, Yuqi Liu, Shunchang Ma, Peng Kang, Deling Li, Xiudong Guan, Jian Chen, Wang Jia","doi":"10.7150/thno.124068","DOIUrl":"10.7150/thno.124068","url":null,"abstract":"<p><p><b>Background:</b> Splicing factors play pivotal roles in mRNA processing and are implicated in tumor progression. The aberrant expression of splicing factors is closely associated with the invasiveness and secretion profiles of pituitary neuroendocrine tumors (PitNETs). In this study, we explored the involvement of splicing factors in PIT1-lineage PitNET progression and assessed the feasibility of targeting the splicing process as a therapeutic approach. <b>Methods:</b> Statistical data on PitNET subtypes were obtained from the National Brain Tumor Registry of China (NBTRC), and gene expression analysis was conducted on 40 clinical samples collected for this study. Transcriptome analysis and RNA immunoprecipitation sequencing (RIP-seq) were utilized to examine FUS-mediated alternative splicing and to identify mRNA binding sites in PitNET cells. Minigene splicing assays were employed to confirm the specific exonic and intronic regions. Additionally, Annexin V/PI assays and JC-1 staining were conducted to evaluate apoptosis. <b>Results:</b> The expression of the splicing factor FUS was elevated in PIT1-lineage PitNETs and was correlated with increased proliferative capacity and reduced apoptosis levels. Transcriptome sequencing revealed that the knockdown of FUS led to extensive exon skipping and activated the p53 pathway. In addition to RIP-seq analysis, these findings suggest that FUS contributes to the inclusion of exon 3 to generate full-length MDM2, a well-established negative regulator of p53. Antisense oligonucleotides (ASOs) specifically designed to target binding sequences on pre-mRNAs effectively disrupted the FUS-mediated splicing process, consequently impeding the progression of PitNETs. <b>Conclusions:</b> Our study elucidated the critical function of FUS as a splicing factor in PitNETs. Furthermore, we illustrated that targeting the splicing mechanism associated with MDM2 could restore p53 levels, thereby impeding the progression of PitNETs. This discovery presents a potentially novel strategy for the clinical management of PIT1-lineage PitNETs.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"3032-3049"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775928/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rationale: Hemodynamic shear stress critically influences atherosclerosis progression, yet the molecular mechanisms linking biomechanical stimuli to endothelial activation and vascular pathology remain poorly understood. While circular RNAs (circRNAs) participate in endothelial mechanotransduction, the role of mechanosensitive small nucleolar RNA (snoRNA)-like circRNA-a unique subclass harboring snoRNA sequences-in atherosclerosis is unexplored. Methods: We characterized sno-circCNOT1 using high-throughput RNA sequencing, RNA interference, immunofluorescence, and co-immunoprecipitation. Functional studies were performed in endothelial cells and ApoE⁻/⁻ mice to assess its role in pyroptosis and atherogenesis. Mechanistic investigations included RNA pull-down, mass spectrometry, and gain- and loss-of-function assays to identify sno-circCNOT1-interacting proteins and downstream signaling. Results: We identified sno-circCNOT1, a circular RNA derived from CNOT1 exon 17 and intron 17, which incorporates snoRNA SNORA50A. Its expression was upregulated by pro-atherogenic interleukin-1β and pathological oscillatory shear stress, but downregulated by laminar shear stress. Functionally, sno-circCNOT1 mediated shear stress-dependent regulation of endothelial pyroptosis and inflammation. Endothelial-specific overexpression of sno-circCNOT1 aggravated atherosclerotic lesion formation in ApoE⁻/⁻ mice. Mechanistically, its snoRNA-like motif was essential for nuclear localization and function. sno-circCNOT1 bound the IF-ROD domain of lamin A/C (LMNA), stabilizing LMNA and facilitating its interaction with the N-terminal domain of methyltransferase-like 14 (METTL14-N), thereby enhancing METTL14 stability. This axis activated NOD-like receptor protein 3 (NLRP3) and amplified endothelial inflammation. Conversely, overexpression of METTL14-N to disrupt this signaling axis attenuates endothelial dysfunction and atherosclerosis progression. Conclusions: sno-circCNOT1 is a mechanosensitive snoRNA-like circRNA that promotes endothelial pyroptosis and atherogenesis via the LMNA/METTL14/NLRP3 axis. METTL14-N offers a protein-based therapeutic approach, positioning this regulatory pathway as a druggable target for atherosclerosis.
{"title":"Mechanosensitive snoRNA-like circular RNA sno-circCNOT1 drives endothelial dysfunction and atherosclerosis.","authors":"Lianru Bi, Yihao Zhu, Ziqi Chen, Yiying Yang, Yanlong Leng, Huijie Wang, Jiajie Pan, Xiaozhe Zhang, Zekai Zeng, Yunjun Liang, Guifu Wu, Wendong Fan","doi":"10.7150/thno.122995","DOIUrl":"10.7150/thno.122995","url":null,"abstract":"<p><p><b>Rationale:</b> Hemodynamic shear stress critically influences atherosclerosis progression, yet the molecular mechanisms linking biomechanical stimuli to endothelial activation and vascular pathology remain poorly understood. While circular RNAs (circRNAs) participate in endothelial mechanotransduction, the role of mechanosensitive small nucleolar RNA (snoRNA)-like circRNA-a unique subclass harboring snoRNA sequences-in atherosclerosis is unexplored. <b>Methods:</b> We characterized sno-circCNOT1 using high-throughput RNA sequencing, RNA interference, immunofluorescence, and co-immunoprecipitation. Functional studies were performed in endothelial cells and <i>ApoE⁻/⁻</i> mice to assess its role in pyroptosis and atherogenesis. Mechanistic investigations included RNA pull-down, mass spectrometry, and gain- and loss-of-function assays to identify sno-circCNOT1-interacting proteins and downstream signaling. <b>Results:</b> We identified sno-circCNOT1, a circular RNA derived from <i>CNOT1</i> exon 17 and intron 17, which incorporates snoRNA SNORA50A. Its expression was upregulated by pro-atherogenic interleukin-1β and pathological oscillatory shear stress, but downregulated by laminar shear stress. Functionally, sno-circCNOT1 mediated shear stress-dependent regulation of endothelial pyroptosis and inflammation. Endothelial-specific overexpression of sno-circCNOT1 aggravated atherosclerotic lesion formation in <i>ApoE⁻/⁻</i> mice. Mechanistically, its snoRNA-like motif was essential for nuclear localization and function. sno-circCNOT1 bound the IF-ROD domain of lamin A/C (LMNA), stabilizing LMNA and facilitating its interaction with the N-terminal domain of methyltransferase-like 14 (METTL14-N), thereby enhancing METTL14 stability. This axis activated NOD-like receptor protein 3 (NLRP3) and amplified endothelial inflammation. Conversely, overexpression of METTL14-N to disrupt this signaling axis attenuates endothelial dysfunction and atherosclerosis progression. <b>Conclusions:</b> sno-circCNOT1 is a mechanosensitive snoRNA-like circRNA that promotes endothelial pyroptosis and atherogenesis via the LMNA/METTL14/NLRP3 axis. METTL14-N offers a protein-based therapeutic approach, positioning this regulatory pathway as a druggable target for atherosclerosis.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"2665-2683"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}