Hyeonho Kim, Gaeun Hyun, Seunghye Kim, Changmo Yu, Young-Gi Hong, Jihyeon Yu, Sangsu Bae, Hyun-Woo Rhee, Jaewon Ko, Ji Won Um
Background: Huntington's disease (HD) is a devastating neurodegenerative disorder caused by CAG repeat expansion in the HTT gene, resulting in a polyglutamine-expanded huntingtin (HTT) protein that forms toxic aggregates. Although heat-shock proteins are known to facilitate the refolding or clearance of misfolded proteins, their precise role in modulating protein aggregation in HD remains unclear. Here, we explore the function of caseinolytic peptidase B (ClpB), a mitochondrial AAA+ ATPase and heat-shock protein, in maintaining proteostasis and synaptic integrity in HD. Methods: We examined how CLPB loss or overexpression in human embryonic kidney 293T (HEK293T) cells impacted the aggregation of wild-type HTT (HTT-Q23) and mutant HTT (HTT-Q79). In parallel, AAV-mediated ClpB knockdown or overexpression was applied to the striatum of HD model mice. and HTT aggregation and inhibitory synaptic alterations were assessed. Aggregate burden was quantified via immunostaining, and inhibitory synapse density was evaluated using VGAT immunohistochemistry and electrophysiological recordings. Results: In HEK293T cells, CLPB knockout led to abnormal aggregation of HTT-Q23 while CLPB overexpression reduced the size of HTT-Q79 aggregates. In the mouse striatum, ClpB knockdown increased HTT-Q23 aggregate numbers and altered HTT-Q79 aggregation morphology, whereas CLPB overexpression restored the density and size of VGAT-positive inhibitory synapses and improved inhibitory synaptic transmission in HD model mice. These effects of CLPB overexpression were associated with a reduced mitochondrial aggregation burden, suggesting that ClpB contributes to mitochondrial protein quality control. Conclusions: These results demonstrate that ClpB regulates both physiological and pathological HTT aggregation and contributes to maintaining inhibitory synaptic integrity. By modulating mitochondrial proteostasis, ClpB acts as a protective factor in HD pathology, highlighting its potential as a therapeutic target for neurodegenerative disorders characterized by protein misfolding.
{"title":"Neuronal mitochondrial disaggregase CLPB ameliorates Huntington's disease pathology in mice.","authors":"Hyeonho Kim, Gaeun Hyun, Seunghye Kim, Changmo Yu, Young-Gi Hong, Jihyeon Yu, Sangsu Bae, Hyun-Woo Rhee, Jaewon Ko, Ji Won Um","doi":"10.7150/thno.122651","DOIUrl":"10.7150/thno.122651","url":null,"abstract":"<p><p><b>Background:</b> Huntington's disease (HD) is a devastating neurodegenerative disorder caused by CAG repeat expansion in the <i>HTT</i> gene, resulting in a polyglutamine-expanded huntingtin (HTT) protein that forms toxic aggregates. Although heat-shock proteins are known to facilitate the refolding or clearance of misfolded proteins, their precise role in modulating protein aggregation in HD remains unclear. Here, we explore the function of caseinolytic peptidase B (ClpB), a mitochondrial AAA+ ATPase and heat-shock protein, in maintaining proteostasis and synaptic integrity in HD. <b>Methods:</b> We examined how CLPB loss or overexpression in human embryonic kidney 293T (HEK293T) cells impacted the aggregation of wild-type HTT (HTT-Q23) and mutant HTT (HTT-Q79). In parallel, AAV-mediated <i>ClpB</i> knockdown or overexpression was applied to the striatum of HD model mice. and HTT aggregation and inhibitory synaptic alterations were assessed. Aggregate burden was quantified via immunostaining, and inhibitory synapse density was evaluated using VGAT immunohistochemistry and electrophysiological recordings. <b>Results:</b> In HEK293T cells, <i>CLPB</i> knockout led to abnormal aggregation of HTT-Q23 while CLPB overexpression reduced the size of HTT-Q79 aggregates. In the mouse striatum, <i>ClpB</i> knockdown increased HTT-Q23 aggregate numbers and altered HTT-Q79 aggregation morphology, whereas CLPB overexpression restored the density and size of VGAT-positive inhibitory synapses and improved inhibitory synaptic transmission in HD model mice. These effects of CLPB overexpression were associated with a reduced mitochondrial aggregation burden, suggesting that ClpB contributes to mitochondrial protein quality control. <b>Conclusions:</b> These results demonstrate that ClpB regulates both physiological and pathological HTT aggregation and contributes to maintaining inhibitory synaptic integrity. By modulating mitochondrial proteostasis, ClpB acts as a protective factor in HD pathology, highlighting its potential as a therapeutic target for neurodegenerative disorders characterized by protein misfolding.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 5","pages":"2388-2404"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12712926/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145805700","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}
Ischemic stroke (IS) is accompanied by high disability and mortality. Thrombolysis and neuroprotection are the predominant therapeutic strategies for IS. However, thrombolytic drugs commonly suffer from hemorrhagic risks and unsatisfactory thrombus-targeting delivery. Additionally, the blood-brain barrier (BBB) presents a significant challenge for the effective delivery of neuroprotective drugs. In recent years, nanodrug delivery systems (nano-DDS) have garnered significant attention for their ability to improve drug efficacy in vivo and facilitate BBB penetration. Specifically, thrombus- and cerebral ischemic lesion-targeted nano-DDS have emerged as a versatile toolbox for the precise treatment of IS. Herein, this paper provides an overview on the latest advancements in nano-DDS for IS therapy, covering conventional nanomedicines, cell membrane-camouflaged biomimetic nano-DDS, and exosome-involved nanotherapeutics, with a particular focus on the potential and application of cutting-edge nano-drug delivery techniques. Finally, we discuss the future perspectives and challenges of nano-DDS in the context of IS treatment.
{"title":"Nanotechnology-donated ischemic stroke therapeutics: evolving strategies from the basic to the cutting-edge.","authors":"Yuting Wang, Jiaxu Xu, Wenxuan Yan, Shenwu Zhang, Cong Luo, Yuequan Wang","doi":"10.7150/thno.127504","DOIUrl":"10.7150/thno.127504","url":null,"abstract":"<p><p>Ischemic stroke (IS) is accompanied by high disability and mortality. Thrombolysis and neuroprotection are the predominant therapeutic strategies for IS. However, thrombolytic drugs commonly suffer from hemorrhagic risks and unsatisfactory thrombus-targeting delivery. Additionally, the blood-brain barrier (BBB) presents a significant challenge for the effective delivery of neuroprotective drugs. In recent years, nanodrug delivery systems (nano-DDS) have garnered significant attention for their ability to improve drug efficacy <i>in vivo</i> and facilitate BBB penetration. Specifically, thrombus- and cerebral ischemic lesion-targeted nano-DDS have emerged as a versatile toolbox for the precise treatment of IS. Herein, this paper provides an overview on the latest advancements in nano-DDS for IS therapy, covering conventional nanomedicines, cell membrane-camouflaged biomimetic nano-DDS, and exosome-involved nanotherapeutics, with a particular focus on the potential and application of cutting-edge nano-drug delivery techniques. Finally, we discuss the future perspectives and challenges of nano-DDS in the context of IS treatment.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"3173-3205"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775947/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934964","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: Neuroblastoma (NB) is a predominant extra-cranial malignancy in childhood, while molecular drivers of its progression and effective treatment strategies have yet to be clarified. Methods: RNA sequencing was performed to identify transcriptional regulators and corresponding target genes. To explore the biological effects and underlying mechanisms of these regulators, a comprehensive methodology was utilized, encompassing chromatin immunoprecipitation, dual-luciferase reporter assay, qRT-PCR, western blot, alongside gene over-expression and silencing techniques, co-immunoprecipitation, and mass spectrometry. The MTT assay, soft agar colony formation, Matrigel invasion, and nude mouse xenograft models were applied to assess oncogenic properties. Patient survival was analyzed using the log-rank test. Results: Armadillo repeat containing 12 (ARMC12) was identified as a MYC-interacting modulator within liquid condensates to up-regulate critical nucleoporin-encoding targets (NUP62/NUP93/NUP98), which promoted nuclear pore complex (NPC) biogenesis to facilitate nuclear trafficking of oncogenic effectors, thereby enhancing invasion and metastasis of NB. As a protein within extracellular vesicles of Malassezia globosa colonizing NB tissues, MGL_0381 also facilitated MYC transactivation via physical interaction to accelerate NPC biogenesis and NB progression. Tioconazole (TCZ) and UU-T02 were identified as efficient inhibitors blocking ARMC12-MYC and MGL_0381-MYC interaction, and synergistically reduced NPC number and aggressive features of NB. High ARMC12, MYC, NUP62, NUP93, or NUP98 levels served as markers of unfavorable patient outcomes in clinical cohorts. Conclusions: These findings collectively demonstrate that dual targeting of AMRC12 and Malassezia globosa disrupts MYC liquid condensates-driven NPC biogenesis during NB progression.
{"title":"Dual targeting of AMRC12 and <i>Malassezia globosa</i> disrupts MYC liquid condensates-driven nuclear pore complex biogenesis in neuroblastoma.","authors":"Anpei Hu, Chunhui Yang, Zhijie Wang, Xiaolin Wang, Xinyue Li, Shunchen Zhou, Bosen Zhao, Jiaying Qu, Xiaojing Wang, Liduan Zheng, Qiangsong Tong","doi":"10.7150/thno.120935","DOIUrl":"10.7150/thno.120935","url":null,"abstract":"<p><p><b>Rationale:</b> Neuroblastoma (NB) is a predominant extra-cranial malignancy in childhood, while molecular drivers of its progression and effective treatment strategies have yet to be clarified. <b>Methods:</b> RNA sequencing was performed to identify transcriptional regulators and corresponding target genes. To explore the biological effects and underlying mechanisms of these regulators, a comprehensive methodology was utilized, encompassing chromatin immunoprecipitation, dual-luciferase reporter assay, qRT-PCR, western blot, alongside gene over-expression and silencing techniques, co-immunoprecipitation, and mass spectrometry. The MTT assay, soft agar colony formation, Matrigel invasion, and nude mouse xenograft models were applied to assess oncogenic properties. Patient survival was analyzed using the log-rank test. <b>Results:</b> Armadillo repeat containing 12 (ARMC12) was identified as a MYC-interacting modulator within liquid condensates to up-regulate critical nucleoporin-encoding targets (<i>NUP62</i>/<i>NUP93</i>/<i>NUP98</i>), which promoted nuclear pore complex (NPC) biogenesis to facilitate nuclear trafficking of oncogenic effectors, thereby enhancing invasion and metastasis of NB. As a protein within extracellular vesicles of <i>Malassezia globosa</i> colonizing NB tissues, MGL_0381 also facilitated MYC transactivation via physical interaction to accelerate NPC biogenesis and NB progression. Tioconazole (TCZ) and UU-T02 were identified as efficient inhibitors blocking ARMC12-MYC and MGL_0381-MYC interaction, and synergistically reduced NPC number and aggressive features of NB. High <i>ARMC12</i>, <i>MYC</i>, <i>NUP62</i>, <i>NUP93</i>, or <i>NUP98</i> levels served as markers of unfavorable patient outcomes in clinical cohorts. <b>Conclusions:</b> These findings collectively demonstrate that dual targeting of AMRC12 and <i>Malassezia globosa</i> disrupts MYC liquid condensates-driven NPC biogenesis during NB progression.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"2866-2886"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775802/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934961","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}
Yuning Liu, Yinyin Li, Hui Zhou, Jieli Yu, Pei Deng, Weiwei Xu, Bingqian Pan, Lei Zhang, Hong Zhou, Jing Zhang, Xiaohan Liu, Min Shi, Xianchi Dong, Bei Tong, Enguang Bi, Nannan Guo, Yu Hu
Rationale: Phospholipase A2 receptor (PLA2R) is the predominant autoantigen in primary membranous nephropathy (PMN), accounting for approximately 70% of clinical cases. However, the mechanisms by which PLA2R initiates and sustains autoimmunity in PMN remain unclear. PLA2R belongs to the mannose receptor (MR) family, members of which have been shown to undergo endocytosis and lysosomal degradation for MHCII-mediated antigen presentation. This study investigates whether antibody binding promotes PLA2R internalization and lysosomal processing to enhance MHCII-mediated antigen presentation and CD4⁺ T cell activation, thereby contributing to the perpetuation of autoimmunity in PMN. Methods: Multiple PLA2R-overexpressing cell lines were generated by lentiviral-mediated overexpression of PLA2R. Imaging and western blot were employed to assess the effects of anti-PLA2R antibodies, derived from PMN patients or produced in-house, on PLA2R internalization and degradation. To define the specific endocytic pathway involved, we used pharmacological inhibitors of endocytosis as well as PLA2R constructs lacking the endocytic domain. Finally, T cell activation was evaluated using OT-II CD4⁺ T cells co-cultured with PLA2R-ovalbumin (OVA)-expressing mouse dendritic cells treated with anti-PLA2R antibodies. Results: Binding of anti-PLA2R antibodies triggers clathrin-mediated endocytosis and lysosomal trafficking of PLA2R. Antibody-induced PLA2R degradation was effectively prevented by specific endocytosis inhibitors or by deletion of the PLA2R endocytic domain. Furthermore, PLA2R-OVA-expressing mouse dendritic cells exposed to PLA2R antibodies enhanced the activation of OVA-specific CD4⁺ T cells both in vitro and in vivo. Conclusions: This study demonstrates that anti-PLA2R antibody induces internalization and lysosomal degradation of PLA2R, a process that may enhance MHC class II-mediated antigen presentation and promote the expansion of antigen-specific CD4⁺ T cells. This mechanism could establish a self-reinforcing feedback loop that perpetuates autoimmune responses in PMN.
{"title":"Antibody-induced internalization and degradation of PLA2R amplifies CD4<sup>+</sup> T cell activation.","authors":"Yuning Liu, Yinyin Li, Hui Zhou, Jieli Yu, Pei Deng, Weiwei Xu, Bingqian Pan, Lei Zhang, Hong Zhou, Jing Zhang, Xiaohan Liu, Min Shi, Xianchi Dong, Bei Tong, Enguang Bi, Nannan Guo, Yu Hu","doi":"10.7150/thno.123035","DOIUrl":"10.7150/thno.123035","url":null,"abstract":"<p><p><b>Rationale:</b> Phospholipase A2 receptor (PLA2R) is the predominant autoantigen in primary membranous nephropathy (PMN), accounting for approximately 70% of clinical cases. However, the mechanisms by which PLA2R initiates and sustains autoimmunity in PMN remain unclear. PLA2R belongs to the mannose receptor (MR) family, members of which have been shown to undergo endocytosis and lysosomal degradation for MHCII-mediated antigen presentation. This study investigates whether antibody binding promotes PLA2R internalization and lysosomal processing to enhance MHCII-mediated antigen presentation and CD4⁺ T cell activation, thereby contributing to the perpetuation of autoimmunity in PMN. <b>Methods:</b> Multiple PLA2R-overexpressing cell lines were generated by lentiviral-mediated overexpression of PLA2R. Imaging and western blot were employed to assess the effects of anti-PLA2R antibodies, derived from PMN patients or produced in-house, on PLA2R internalization and degradation. To define the specific endocytic pathway involved, we used pharmacological inhibitors of endocytosis as well as PLA2R constructs lacking the endocytic domain. Finally, T cell activation was evaluated using OT-II CD4⁺ T cells co-cultured with PLA2R-ovalbumin (OVA)-expressing mouse dendritic cells treated with anti-PLA2R antibodies. <b>Results:</b> Binding of anti-PLA2R antibodies triggers clathrin-mediated endocytosis and lysosomal trafficking of PLA2R. Antibody-induced PLA2R degradation was effectively prevented by specific endocytosis inhibitors or by deletion of the PLA2R endocytic domain. Furthermore, PLA2R-OVA-expressing mouse dendritic cells exposed to PLA2R antibodies enhanced the activation of OVA-specific CD4⁺ T cells both <i>in vitro</i> and <i>in vivo</i>. <b>Conclusions:</b> This study demonstrates that anti-PLA2R antibody induces internalization and lysosomal degradation of PLA2R, a process that may enhance MHC class II-mediated antigen presentation and promote the expansion of antigen-specific CD4⁺ T cells. This mechanism could establish a self-reinforcing feedback loop that perpetuates autoimmune responses in PMN.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 1","pages":"37-57"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12665110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145655723","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: In ulcerative colitis (UC), microbial products or metabolites, coupled with inflammatory stimuli, result in simultaneous damage to both the intestinal epithelial barrier (IEB) and gut vascular barrier (GVB). Current UC treatments usually focus on modulating IEB, whereas GVB-which critically regulates the translocation of gut microbiota and metabolites into systemic circulation-has been largely overlooked. Here, we developed a facile, biomimetic strategy to engineer anti-inflammatory berberine/magnolol self-assembled nanoparticles (BM NPs) using macrophage membrane camouflage, enabling targeted UC accumulation and dual restoration of both the IEB and GVB. Methods: BM NPs employing macrophage membranes to camouflage mimetic nanoplatform. The mimetic nanoplatform on targeting capacity of inflamed intestinal epithelial cells, M1/M2 polarization, macrophage and intestinal epithelial cell inflammatory factors, and vascular endothelial cell migration and tube-forming were evaluated in vitro. Furthermore, its therapeutic efficacy was assessed in a mice UC model, demonstrating significant reductions in bacterial translocation, restoration of both the IEB and GVB, and modulation of the inflammatory immune microenvironment. Results: The biomimetic nanoplatform demonstrates superior targeting specificity and prolonged retention in inflamed intestinal epithelium and vascular tissues. Macrophage membranes achieve GVB repair by mechanical traction and physical adsorption of inflammatory factors. Besides, efficient delivery of the loaded anti-inflammatory drugs also achieves the repair of the IEB. GVB repair effectively prevents systemic dissemination of gut-derived microbes and their metabolites, thereby attenuating UC-induced inflammatory cascades. Collectively, this approach significantly ameliorates colonic pathology in UC. Conclusion: Our study proposes the synergistic repair of IEB and GVB through the mechanical traction and physical adsorption of macrophage membranes, assisted by anti-inflammatory components, which provide new insights as well as a new paradigm for the treatment of UC.
{"title":"Multifunctional macrophage mimetic nanoplatform modulates vascular and epithelial double gut barriers to alleviate ulcerative colitis.","authors":"Weijian Cheng, Yixi Zhu, Miaoxizi Luo, Xiao Wang, Quanlong Chen, Siyao Li, Jing Xian, Meng Xiao, Licheng Liu, Yuanyuan Wang, Chaomei Fu, Ruibing Wang, Qian Cheng, Jinming Zhang","doi":"10.7150/thno.118236","DOIUrl":"10.7150/thno.118236","url":null,"abstract":"<p><p><b>Rationale:</b> In ulcerative colitis (UC), microbial products or metabolites, coupled with inflammatory stimuli, result in simultaneous damage to both the intestinal epithelial barrier (IEB) and gut vascular barrier (GVB). Current UC treatments usually focus on modulating IEB, whereas GVB-which critically regulates the translocation of gut microbiota and metabolites into systemic circulation-has been largely overlooked. Here, we developed a facile, biomimetic strategy to engineer anti-inflammatory berberine/magnolol self-assembled nanoparticles (BM NPs) using macrophage membrane camouflage, enabling targeted UC accumulation and dual restoration of both the IEB and GVB. <b>Methods:</b> BM NPs employing macrophage membranes to camouflage mimetic nanoplatform. The mimetic nanoplatform on targeting capacity of inflamed intestinal epithelial cells, M1/M2 polarization, macrophage and intestinal epithelial cell inflammatory factors, and vascular endothelial cell migration and tube-forming were evaluated <i>in vitro</i>. Furthermore, its therapeutic efficacy was assessed in a mice UC model, demonstrating significant reductions in bacterial translocation, restoration of both the IEB and GVB, and modulation of the inflammatory immune microenvironment. <b>Results:</b> The biomimetic nanoplatform demonstrates superior targeting specificity and prolonged retention in inflamed intestinal epithelium and vascular tissues. Macrophage membranes achieve GVB repair by mechanical traction and physical adsorption of inflammatory factors. Besides, efficient delivery of the loaded anti-inflammatory drugs also achieves the repair of the IEB. GVB repair effectively prevents systemic dissemination of gut-derived microbes and their metabolites, thereby attenuating UC-induced inflammatory cascades. Collectively, this approach significantly ameliorates colonic pathology in UC. <b>Conclusion:</b> Our study proposes the synergistic repair of IEB and GVB through the mechanical traction and physical adsorption of macrophage membranes, assisted by anti-inflammatory components, which provide new insights as well as a new paradigm for the treatment of UC.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 1","pages":"58-76"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12665101/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145655376","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}
Endometrial regeneration remains a significant clinical challenge for women with intrauterine adhesions (IUAs), thin endometrium, or uterine factor infertility, conditions that severely impair fertility and reproductive outcomes. Traditional hormonal and surgical interventions often fail to restore the structural and functional integrity of damaged endometrial tissue. This review comprehensively examines integrative bioengineering strategies for endometrial regeneration, focusing on the synergistic applications of biomaterials, stem cells, organoids, and organ-on-a-chip technologies. Natural polymers such as collagen, gelatin, alginate, hyaluronic acid, and synthetic polymers including PCL, PLA, PGA, and PLGA have been comprehensively evaluated for their ability to mimic extracellular matrix, support cell proliferation, angiogenesis, and modulate immune responses. The incorporation of mesenchymal stem cells, extracellular vesicles, and growth factors into bioengineered scaffolds, such as hydrogels and nanofiber membranes, enhances regenerative efficacy. Furthermore, emerging platforms, such as endometrial organoids, 3D bioprinting, and organ-on-a-chip systems, offer physiologically relevant models for precision regenerative medicine. Innovations such as AI-assisted monitoring, 4D printing, and advanced drug delivery systems represent transformative approaches to overcome current therapeutic limitations. This review highlights the convergence of materials science, stem cell biology, and microengineering as a foundation for next-generation, personalized therapies aimed at restoring endometrial function and fertility. In addition, the review highlights biomaterial-based strategies as the foundation of endometrial regeneration, by detailing how natural polymers (e.g., collagen, gelatin, alginate, hyaluronic acid) and synthetic polymers (e.g., PCL, PLA, PLGA) support tissue repair structurally and by mediating biological functions. The integration of advanced technologies, such as 4D printing, AI-assisted monitoring, and stem cell-derived extracellular vesicle delivery has emerged as a transformative direction for overcoming current clinical challenges. Collectively, these approaches offer a next-generation therapeutic paradigm for restoring endometrial function and fertility.
{"title":"Integrative bioengineering strategies for endometrial regeneration: From biomaterials and stem cells to organoids and organ-on-a-chip technologies.","authors":"Soo-Rim Kim, Hwa-Yong Lee","doi":"10.7150/thno.123298","DOIUrl":"10.7150/thno.123298","url":null,"abstract":"<p><p>Endometrial regeneration remains a significant clinical challenge for women with intrauterine adhesions (IUAs), thin endometrium, or uterine factor infertility, conditions that severely impair fertility and reproductive outcomes. Traditional hormonal and surgical interventions often fail to restore the structural and functional integrity of damaged endometrial tissue. This review comprehensively examines integrative bioengineering strategies for endometrial regeneration, focusing on the synergistic applications of biomaterials, stem cells, organoids, and organ-on-a-chip technologies. Natural polymers such as collagen, gelatin, alginate, hyaluronic acid, and synthetic polymers including PCL, PLA, PGA, and PLGA have been comprehensively evaluated for their ability to mimic extracellular matrix, support cell proliferation, angiogenesis, and modulate immune responses. The incorporation of mesenchymal stem cells, extracellular vesicles, and growth factors into bioengineered scaffolds, such as hydrogels and nanofiber membranes, enhances regenerative efficacy. Furthermore, emerging platforms, such as endometrial organoids, 3D bioprinting, and organ-on-a-chip systems, offer physiologically relevant models for precision regenerative medicine. Innovations such as AI-assisted monitoring, 4D printing, and advanced drug delivery systems represent transformative approaches to overcome current therapeutic limitations. This review highlights the convergence of materials science, stem cell biology, and microengineering as a foundation for next-generation, personalized therapies aimed at restoring endometrial function and fertility. In addition, the review highlights biomaterial-based strategies as the foundation of endometrial regeneration, by detailing how natural polymers (e.g., collagen, gelatin, alginate, hyaluronic acid) and synthetic polymers (e.g., PCL, PLA, PLGA) support tissue repair structurally and by mediating biological functions. The integration of advanced technologies, such as 4D printing, AI-assisted monitoring, and stem cell-derived extracellular vesicle delivery has emerged as a transformative direction for overcoming current clinical challenges. Collectively, these approaches offer a next-generation therapeutic paradigm for restoring endometrial function and fertility.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"736-775"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674934/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678813","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}
Sujin Kim, Suyeon Lee, Manseok Yang, Seungwon Jung, Nanhee Song, Nuri Kim, Hanui Jo, Seunga Lee, Chaihong Nah, Seong-Cheol Park, Dongwon Lee
Rationale:Gamma-glutamyl transferase (GGT) is overexpressed on cancer cell membranes and has been widely used as a promising target for receptor-mediated therapy. However, its heterogeneous expression limits targeting efficacy. Based on the notation that reactive oxygen species (ROS) upregulate GGT and induce oxidative stress-mediated cancer cell death, we hypothesized that GGT-targeted ROS generation could simultaneously induce cell death and also reprogram tumors to achieve self-boosting targeted therapy. Methods: We developed GLOXmp, a glutamic acid (Glu)-coated oxidative stress nanoamplifier, in which glutathione (GSH)-depleting B2C was loaded in ROS-generating amphiphilic polyCA. GLOXmp was designed to induce oxidative stress, modulate GGT expression, subsequently enhancing tumor targeting both in vitro and in vivo using xenograft mouse models. Results: GLOXmp internalized GGT-overexpressing cancer cells and concurrently generated ROS and depleted intracellular GSH, leading to mitochondrial damage and potent cancer cell death. Importantly, GLOXmp reprogrammed tumor cells to upregulate GGT, leading to the enhancement of receptor-mediated uptake of subsequent doses. In tumor xenograft model, repeated administration of GLOXmp significantly elevated oxidative stress, increased GGT expression, and effectively eradicated tumors without systemic toxicity. Conclusion: GLOXmp specifically targeted GGT-overexpressing cancer cells and effectively suppressed tumor development through oxidative stress amplification. Given a self-reinforcing strategy for targeted cancer therapy through oxidative stress-mediated tumor cell reprogramming, GLOXmp demonstrates represents a promising advancement in precision nanomedicine.
{"title":"Self-boosting targeted anticancer therapy <i>via</i> cancer cell self-reprogramming with GGT-targeting oxidative stress nanoamplifiers.","authors":"Sujin Kim, Suyeon Lee, Manseok Yang, Seungwon Jung, Nanhee Song, Nuri Kim, Hanui Jo, Seunga Lee, Chaihong Nah, Seong-Cheol Park, Dongwon Lee","doi":"10.7150/thno.120954","DOIUrl":"10.7150/thno.120954","url":null,"abstract":"<p><p><b>Rationale:</b> <i>Gamma</i>-glutamyl transferase (GGT) is overexpressed on cancer cell membranes and has been widely used as a promising target for receptor-mediated therapy. However, its heterogeneous expression limits targeting efficacy. Based on the notation that reactive oxygen species (ROS) upregulate GGT and induce oxidative stress-mediated cancer cell death, we hypothesized that GGT-targeted ROS generation could simultaneously induce cell death and also reprogram tumors to achieve self-boosting targeted therapy. <b>Methods:</b> We developed GLOXmp, a glutamic acid (Glu)-coated oxidative stress nanoamplifier, in which glutathione (GSH)-depleting B2C was loaded in ROS-generating amphiphilic polyCA. GLOXmp was designed to induce oxidative stress, modulate GGT expression, subsequently enhancing tumor targeting both in vitro and in vivo using xenograft mouse models. <b>Results:</b> GLOXmp internalized GGT-overexpressing cancer cells and concurrently generated ROS and depleted intracellular GSH, leading to mitochondrial damage and potent cancer cell death. Importantly, GLOXmp reprogrammed tumor cells to upregulate GGT, leading to the enhancement of receptor-mediated uptake of subsequent doses. In tumor xenograft model, repeated administration of GLOXmp significantly elevated oxidative stress, increased GGT expression, and effectively eradicated tumors without systemic toxicity. <b>Conclusion:</b> GLOXmp specifically targeted GGT-overexpressing cancer cells and effectively suppressed tumor development through oxidative stress amplification. Given a self-reinforcing strategy for targeted cancer therapy through oxidative stress-mediated tumor cell reprogramming, GLOXmp demonstrates represents a promising advancement in precision nanomedicine.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 4","pages":"1925-1940"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12680591/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701631","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}
Audrey Picot, Charlène Jacqmarcq, Célia Seillier, Sara Martinez de Lizarrondo, Maxime Gauberti, Ankita Talukdar, Igor Khalin, Clarisse Mouriaux, Pierre Mangin, Didier Goux, Peter Schmidt, Denis Vivien, Thomas Bonnard
Background: Microthrombi obstructing downstream microcirculation in acute ischemic stroke (AIS) are difficult to treat and visualize with current imaging methods. Methods: To address this need, a novel theranostic agent, IO@PDA@tPA, was developed by combining iron oxide microparticles (IO) coated with polydopamine (PDA) and conjugated with recombinant tissue-type plasminogen activator (r-tPA). The amidolytic and fibrinolytic capacities of r-tPA grafted on IO@PDA were assessed using the spectrofluorometric test, the clot lysis assay, and the whole blood halo assay. IO@PDA@tPA was then tested in vivo in a preclinical ischemic stroke model induced by thrombin injection into the middle cerebral artery in both non-diabetic and diabetic mice. Two doses equivalent to 2.5 and 5 mg/kg r-tPA were tested. The presence of microthrombi was monitored via molecular MRI. A series of T2*-weighted sequences for microthrombi imaging and magnetic resonance angiography (MRA) was performed over 45 min. At 24 h, lesion size, vessel patency, and hemorrhagic transformation were assessed with T2 -weighted imaging, MRA, and T2* -weighted MRI, respectively. A grip test was performed to assess functional recovery one day before stroke (baseline), and at 24 h and five days after stroke. Additionally, inflammatory processes were evaluated five days post-stroke by flow cytometry in the non-diabetic cohort. Results: This agent exhibited in vitro clot lysis activity. In vivo, administration of IO@PDA@tPA at one-quarter of the standard r-tPA dose enabled both visualization and degradation of microthrombi, as detected by T2* -weighted MRI. This treatment significantly reduced lesion size and promoted recanalization 24 h after stroke onset. In the hyperglycemic mice cohort, the agent demonstrated efficacy comparable to r-tPA without increasing hemorrhagic risk-a common complication of free r-tPA. Moreover, full functional recovery observed within five days post-stroke. Flow cytometry indicated that IO@PDA@tPA mitigated inflammatory processes. Conclusion: IO@PDA@tPA represents a promising theranostic agent targeting microthrombi in AIS, reducing the required r-tPA dose and limiting associated side effects.
{"title":"Engineered nanoconjugates for simultaneous detection and degradation of stroke-associated microthrombi.","authors":"Audrey Picot, Charlène Jacqmarcq, Célia Seillier, Sara Martinez de Lizarrondo, Maxime Gauberti, Ankita Talukdar, Igor Khalin, Clarisse Mouriaux, Pierre Mangin, Didier Goux, Peter Schmidt, Denis Vivien, Thomas Bonnard","doi":"10.7150/thno.119705","DOIUrl":"10.7150/thno.119705","url":null,"abstract":"<p><p><b>Background:</b> Microthrombi obstructing downstream microcirculation in acute ischemic stroke (AIS) are difficult to treat and visualize with current imaging methods. <b>Methods:</b> To address this need, a novel theranostic agent, IO@PDA@tPA, was developed by combining iron oxide microparticles (IO) coated with polydopamine (PDA) and conjugated with recombinant tissue-type plasminogen activator (r-tPA). The amidolytic and fibrinolytic capacities of r-tPA grafted on IO@PDA were assessed using the spectrofluorometric test, the clot lysis assay, and the whole blood halo assay. IO@PDA@tPA was then tested <i>in vivo</i> in a preclinical ischemic stroke model induced by thrombin injection into the middle cerebral artery in both non-diabetic and diabetic mice. Two doses equivalent to 2.5 and 5 mg/kg r-tPA were tested. The presence of microthrombi was monitored via molecular MRI. A series of <i>T</i> <sub>2</sub>*-weighted sequences for microthrombi imaging and magnetic resonance angiography (MRA) was performed over 45 min. At 24 h, lesion size, vessel patency, and hemorrhagic transformation were assessed with <i>T<sub>2</sub></i> -weighted imaging, MRA, and <i>T<sub>2</sub> <sup>*</sup></i> -weighted MRI, respectively. A grip test was performed to assess functional recovery one day before stroke (baseline), and at 24 h and five days after stroke. Additionally, inflammatory processes were evaluated five days post-stroke by flow cytometry in the non-diabetic cohort. <b>Results:</b> This agent exhibited <i>in vitro</i> clot lysis activity. <i>In vivo</i>, administration of IO@PDA@tPA at one-quarter of the standard r-tPA dose enabled both visualization and degradation of microthrombi, as detected by <i>T<sub>2</sub> <sup>*</sup></i> -weighted MRI. This treatment significantly reduced lesion size and promoted recanalization 24 h after stroke onset. In the hyperglycemic mice cohort, the agent demonstrated efficacy comparable to r-tPA without increasing hemorrhagic risk-a common complication of free r-tPA. Moreover, full functional recovery observed within five days post-stroke. Flow cytometry indicated that IO@PDA@tPA mitigated inflammatory processes. <b>Conclusion:</b> IO@PDA@tPA represents a promising theranostic agent targeting microthrombi in AIS, reducing the required r-tPA dose and limiting associated side effects.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 3","pages":"1082-1102"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679082/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701567","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}
Ji Yong Park, Jinyeong Choi, Jeongbin Park, Jin Sil Kim, Young Chan Ann, Hyewon Chung, Jisu Park, Jiyoon Kim, Seung Hyeok Seok, Hongyoon Choi, Hyung-Jun Im, Yun-Sang Lee
Rationale: Glycosylation of drug delivery vehicles enables selective tumor microenvironment (TME) targeting but is limited by the lack of precise glycan control and unbiased evaluation of in situ targeting. We developed a clickable albumin nanoplatform engineered by distinct glycosylation for selective in vivo cell targeting (CAN-DGIT) with a defined number of sugar moieties and integrated spatial transcriptomics (ST) to map nanoparticle-TME interactions. Methods: Albumin was functionalized with azadibenzocyclooctyne (ADIBO) at a controlled degree of functionalization (DOF), confirmed by MALDI-TOF and UV-vis spectroscopy, followed by conjugation of azide-functionalized mannose, galactose, or glucose via click chemistry. Nanoparticles were labeled with 64Cu or fluorescent dyes for PET imaging and ex vivo analysis in healthy and 4T1 tumor-bearing mice. ST based algorithms, spatial gene-image integration (SPADE), cell-type deconvolution (CellDART), and image-based molecular signature analysis (IAMSAM), were used to define TME clusters, associated cell populations, and glycan receptor gene signatures. Clodronate-loaded glycosylated albumins were tested for tumor-associated macrophage (TAM) depletion. Results: Glycosylation type of CAN-DGIT dictated pharmacokinetics and targeting. Mannosylated albumin (Man-Alb) showed rapid hepatic retention via mannose receptors on Kupffer cells and TAMs; galactosylated albumin (Gal-Alb) exhibited rapid hepatobiliary clearance with the highest tumor-to-liver ratio; glucosylated albumin at the C6 position (Glc(6)-Alb) progressively accumulated in tumors, correlating with glucose transporter 1 (GLUT1)-expressing cancer cells. ST confirmed Man-Alb enrichment in extracellular matrix (ECM)/TAM-rich clusters (mannose receptor C-type 1, Mrc1-high) and Gal-/Glc-Alb uptake in glycolytic/hypoxic tumor clusters (Slc2a1-high). Man-Alb-clodronate achieved potent CD206+ TAM depletion without altering drug release kinetics. Conclusions: Precisely tuned glycosylation enables programmable biodistribution and cell-type targeting of albumin nanoparticles in the TME. Integrating PET with ST provides a robust framework for mechanistic mapping of nanomedicine uptake. The CAN-DGIT platform offers a versatile strategy for developing targeted theranostic agents with immunomodulatory potential.
{"title":"Engineering and evaluation of precision-glycosylated clickable albumin nanoplatform for targeting the tumor microenvironment.","authors":"Ji Yong Park, Jinyeong Choi, Jeongbin Park, Jin Sil Kim, Young Chan Ann, Hyewon Chung, Jisu Park, Jiyoon Kim, Seung Hyeok Seok, Hongyoon Choi, Hyung-Jun Im, Yun-Sang Lee","doi":"10.7150/thno.123973","DOIUrl":"10.7150/thno.123973","url":null,"abstract":"<p><p><b>Rationale:</b> Glycosylation of drug delivery vehicles enables selective tumor microenvironment (TME) targeting but is limited by the lack of precise glycan control and unbiased evaluation of in situ targeting. We developed a clickable albumin nanoplatform engineered by distinct glycosylation for selective <i>in vivo</i> cell targeting (CAN-DGIT) with a defined number of sugar moieties and integrated spatial transcriptomics (ST) to map nanoparticle-TME interactions. <b>Methods</b>: Albumin was functionalized with azadibenzocyclooctyne (ADIBO) at a controlled degree of functionalization (DOF), confirmed by MALDI-TOF and UV-vis spectroscopy, followed by conjugation of azide-functionalized mannose, galactose, or glucose via click chemistry. Nanoparticles were labeled with <sup>64</sup>Cu or fluorescent dyes for PET imaging and <i>ex vivo</i> analysis in healthy and 4T1 tumor-bearing mice. ST based algorithms, spatial gene-image integration (SPADE), cell-type deconvolution (CellDART), and image-based molecular signature analysis (IAMSAM), were used to define TME clusters, associated cell populations, and glycan receptor gene signatures. Clodronate-loaded glycosylated albumins were tested for tumor-associated macrophage (TAM) depletion. <b>Results:</b> Glycosylation type of CAN-DGIT dictated pharmacokinetics and targeting. Mannosylated albumin (Man-Alb) showed rapid hepatic retention via mannose receptors on Kupffer cells and TAMs; galactosylated albumin (Gal-Alb) exhibited rapid hepatobiliary clearance with the highest tumor-to-liver ratio; glucosylated albumin at the C6 position (Glc(6)-Alb) progressively accumulated in tumors, correlating with glucose transporter 1 (GLUT1)-expressing cancer cells. ST confirmed Man-Alb enrichment in extracellular matrix (ECM)/TAM-rich clusters (mannose receptor C-type 1, Mrc1-high) and Gal-/Glc-Alb uptake in glycolytic/hypoxic tumor clusters (Slc2a1-high). Man-Alb-clodronate achieved potent CD206+ TAM depletion without altering drug release kinetics. <b>Conclusions:</b> Precisely tuned glycosylation enables programmable biodistribution and cell-type targeting of albumin nanoparticles in the TME. Integrating PET with ST provides a robust framework for mechanistic mapping of nanomedicine uptake. The CAN-DGIT platform offers a versatile strategy for developing targeted theranostic agents with immunomodulatory potential.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 3","pages":"1482-1503"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701593","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}
Haijun Li, Xianzhou Huang, Jin Yang, Liping Bai, Meiling Shen, Yaqin Zhao, Changyang Gong, Yanjie You, Qinjie Wu
Background: Radiotherapy (RT)-induced antitumor immunity has attracted extensive attention. However, such antitumor immunity often proves inadequate to combat metastatic and recurrent tumors in clinical practice. While hypoxia severely limits the initiation of adequate systemic antitumor immune responses, the strength of such responses is further compromised by the blockage of effector T cells, ultimately undermining the ability of RT to elicit a robust abscopal effect and durable immune memory. Methods: Hence, a limitation-circumventing strength-capitalizing hydrogel (UP) is developed to increase the efficacy of RT for a robust abscopal effect and durable antitumor immunity to cope with metastatic and recurrent tumors. Results: With single-administration and radiation treatments, UP efficiently generates oxygen in situ to circumvent the limitation of RT hypoxia. After such limitations are overcome, the tumor-killing capacity of RT is significantly promoted, resulting in strong antitumor immune responses. Moreover, UP further inhibits immune checkpoints to reinvigorate effector T cells, capitalizing on the strength of RT-induced antitumor immune responses. With such strength-capitalization, RT-induced transient immune responses are amplified to systemic immunity, triggering a robust abscopal effect to eliminate distant metastasis and establishing durable immune memory against recurrence. Conclusions: Consequently, UP potentiates the antitumor immunity of RT through circumventing the hypoxia barrier and capitalizing on immune activation. Our study provides new insights into RT enhancement.
{"title":"Limitation-circumventing and strength-capitalizing hydrogel potentiates durable antitumor immunity and robust abscopal effect in radiotherapy.","authors":"Haijun Li, Xianzhou Huang, Jin Yang, Liping Bai, Meiling Shen, Yaqin Zhao, Changyang Gong, Yanjie You, Qinjie Wu","doi":"10.7150/thno.118974","DOIUrl":"10.7150/thno.118974","url":null,"abstract":"<p><p><b>Background:</b> Radiotherapy (RT)-induced antitumor immunity has attracted extensive attention. However, such antitumor immunity often proves inadequate to combat metastatic and recurrent tumors in clinical practice. While hypoxia severely limits the initiation of adequate systemic antitumor immune responses, the strength of such responses is further compromised by the blockage of effector T cells, ultimately undermining the ability of RT to elicit a robust abscopal effect and durable immune memory. <b>Methods:</b> Hence, a limitation-circumventing strength-capitalizing hydrogel (UP) is developed to increase the efficacy of RT for a robust abscopal effect and durable antitumor immunity to cope with metastatic and recurrent tumors. <b>Results:</b> With single-administration and radiation treatments, UP efficiently generates oxygen <i>in situ</i> to circumvent the limitation of RT hypoxia. After such limitations are overcome, the tumor-killing capacity of RT is significantly promoted, resulting in strong antitumor immune responses. Moreover, UP further inhibits immune checkpoints to reinvigorate effector T cells, capitalizing on the strength of RT-induced antitumor immune responses. With such strength-capitalization, RT-induced transient immune responses are amplified to systemic immunity, triggering a robust abscopal effect to eliminate distant metastasis and establishing durable immune memory against recurrence. <b>Conclusions:</b> Consequently, UP potentiates the antitumor immunity of RT through circumventing the hypoxia barrier and capitalizing on immune activation. Our study provides new insights into RT enhancement.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 3","pages":"1432-1444"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12679582/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145701600","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}