Jiawen Chen, Junfeng Liu, Xiaoting Zhang, Xi Li, Jinming Fan, Shengchao Zhao, Junbin Liu, Bin Zhou, Ke Zhang
Background: Hepatocellular carcinoma (HCC), the major form of primary liver cancer, contributes markedly to cancer-related mortality worldwide and remains a serious global health concern, particularly affecting individuals with underlying chronic liver disorders. In hepatocellular carcinoma, insufficient radiofrequency ablation (iRFA) has been reported to drive local tumor relapse and distant spread, possibly by aggravating the immunosuppressive features of the tumor microenvironment. The present work seeks to clarify the underlying pathways driving the development of an immunosuppressive milieu after RFA and to identify potential therapeutic approaches to counteract this process. Methods: An injectable hydrogel composed of quaternized chitosan (QCS) and tannic acid (TA) was constructed to encapsulate verteporfin (VP), a well-established photosensitizer that has been clinically applied for treating neovascular retinal disorders such as age-related macular disease. Beyond its ophthalmologic application, VP has recently been reported to display anti-tumor activity through inhibition of oncogenic regulators such as Yes-associated protein (YAP), indicating its potential utility in cancer therapy. This hydrogel formulation is designed to target residual tumor tissue post-RFA, providing localized delivery and sustained release of VP to enhance anti-tumor immune responses. Results: Our findings identified YAP activation as a critical mediator of immunosuppression in residual tumors following RFA. Pharmacological inhibition of YAP significantly reduced the infiltration of myeloid-derived suppressor cells (MDSCs) and effectively reversed the immunosuppressive microenvironment conditions. Furthermore, the QCS/TA hydrogel enabled sustained local release of VP, resulting in enhanced antitumor immune responses via MDSC suppression. When administered as an adjuvant therapy following suboptimal RFA, the hydrogel markedly inhibited the progression of residual tumors, highlighting its therapeutic potential in improving post-RFA outcomes. Conclusion: Collectively, our data suggest YAP pathway inhibition as a promising immunomodulatory strategy to complement RFA in HCC management. This work demonstrates that the QCS/TA hydrogel-based delivery system can remodel the tumor immune milieu to overcome immunosuppression and delay post-ablation tumor recurrence, supporting its potential as a translational drug delivery strategy.
{"title":"Verteporfin-loaded hydrogel targeting YAP-mediated MDSCs recruitment for the treatment of residual tumors after incomplete radiofrequency ablation.","authors":"Jiawen Chen, Junfeng Liu, Xiaoting Zhang, Xi Li, Jinming Fan, Shengchao Zhao, Junbin Liu, Bin Zhou, Ke Zhang","doi":"10.7150/thno.119377","DOIUrl":"10.7150/thno.119377","url":null,"abstract":"<p><p><b>Background:</b> Hepatocellular carcinoma (HCC), the major form of primary liver cancer, contributes markedly to cancer-related mortality worldwide and remains a serious global health concern, particularly affecting individuals with underlying chronic liver disorders. In hepatocellular carcinoma, insufficient radiofrequency ablation (iRFA) has been reported to drive local tumor relapse and distant spread, possibly by aggravating the immunosuppressive features of the tumor microenvironment. The present work seeks to clarify the underlying pathways driving the development of an immunosuppressive milieu after RFA and to identify potential therapeutic approaches to counteract this process. <b>Methods:</b> An injectable hydrogel composed of quaternized chitosan (QCS) and tannic acid (TA) was constructed to encapsulate verteporfin (VP), a well-established photosensitizer that has been clinically applied for treating neovascular retinal disorders such as age-related macular disease. Beyond its ophthalmologic application, VP has recently been reported to display anti-tumor activity through inhibition of oncogenic regulators such as Yes-associated protein (YAP), indicating its potential utility in cancer therapy. This hydrogel formulation is designed to target residual tumor tissue post-RFA, providing localized delivery and sustained release of VP to enhance anti-tumor immune responses. <b>Results:</b> Our findings identified YAP activation as a critical mediator of immunosuppression in residual tumors following RFA. Pharmacological inhibition of YAP significantly reduced the infiltration of myeloid-derived suppressor cells (MDSCs) and effectively reversed the immunosuppressive microenvironment conditions. Furthermore, the QCS/TA hydrogel enabled sustained local release of VP, resulting in enhanced antitumor immune responses via MDSC suppression. When administered as an adjuvant therapy following suboptimal RFA, the hydrogel markedly inhibited the progression of residual tumors, highlighting its therapeutic potential in improving post-RFA outcomes. <b>Conclusion:</b> Collectively, our data suggest YAP pathway inhibition as a promising immunomodulatory strategy to complement RFA in HCC management. This work demonstrates that the QCS/TA hydrogel-based delivery system can remodel the tumor immune milieu to overcome immunosuppression and delay post-ablation tumor recurrence, supporting its potential as a translational drug delivery strategy.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 5","pages":"2372-2387"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12712912/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145805349","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: Depression is a heterogeneous disorder influenced by cell type-specific gene transcription in the brain. Peroxisome proliferator-activated receptor gamma (PPARγ) plays an important role in modulating the pathophysiology of depression. However, the role of PPARγ signaling in modulating depression-responsive neuronal ensembles remains largely unknown. Methods: We established a chronic restraint stress mouse model and integrated multi-omics and functional approaches to investigate the role of glucosylceramide (GlcCer)-PPARγ signaling in stress-induced depression. Conditional knockout mice targeting glucosylceramide synthase (GCS) or Pparg in dopamine D2 receptor-expressing medium spiny neurons (D2-MSNs) were generated using a Cre-loxP system, and molecular assays were used to characterize GlcCer as an endogenous activator of PPARγ-driven transcriptional programs. Results: GlcCer as a crucial native activator of PPARγ that specifically modulates depression by binding to the activation function 1 domain of PPARγ in D2-MSNs in the dorsal striatum. Genetic ablation of GCS in D2-MSNs disrupts PPARγ signaling and neuronal function, leading to depression-like behaviors in mice. Selective deletion of Pparg in D2-MSNs produces a similar effect through dopamine D2 receptor. Administration of GlcCer or the PPARγ agonist pioglitazone reverses stress-induced depression-like behaviors, combined GlcCer and pioglitazone exerts additive antidepressant effects. Conclusions: These findings demonstrate a pivotal role for GlcCer-PPARγ signaling in D2-MSNs in depression, highlighting the therapeutic potential of targeting PPARγ activity in depression.
{"title":"Glucosylceramide regulates depression through activating peroxisome proliferator-activated receptor gamma in dorsal striatum.","authors":"Linhong Jiang, Yuman He, Haxiaoyu Liu, Dingwen Zhang, Yanping Dai, Qian Bu, Quanshan Shi, Huaichuan Duan, Ying Zhao, Shu Li, Shuang Han, Yuanyi Zhou, Yue Zhao, Feng Qin, Yaxing Chen, Liang Wang, Hongchun Li, Chunqi Liu, Meng Qin, Weihong Kuang, Ni Zhang, Yinglan Zhao, Xiaobo Cen","doi":"10.7150/thno.123178","DOIUrl":"10.7150/thno.123178","url":null,"abstract":"<p><p><b>Rationale:</b> Depression is a heterogeneous disorder influenced by cell type-specific gene transcription in the brain. Peroxisome proliferator-activated receptor gamma (PPARγ) plays an important role in modulating the pathophysiology of depression. However, the role of PPARγ signaling in modulating depression-responsive neuronal ensembles remains largely unknown. <b>Methods:</b> We established a chronic restraint stress mouse model and integrated multi-omics and functional approaches to investigate the role of glucosylceramide (GlcCer)-PPARγ signaling in stress-induced depression. Conditional knockout mice targeting glucosylceramide synthase (GCS) or <i>Pparg</i> in dopamine D2 receptor-expressing medium spiny neurons (D2-MSNs) were generated using a Cre-loxP system, and molecular assays were used to characterize GlcCer as an endogenous activator of PPARγ-driven transcriptional programs. <b>Results:</b> GlcCer as a crucial native activator of PPARγ that specifically modulates depression by binding to the activation function 1 domain of PPARγ in D2-MSNs in the dorsal striatum. Genetic ablation of GCS in D2-MSNs disrupts PPARγ signaling and neuronal function, leading to depression-like behaviors in mice. Selective deletion of <i>Pparg</i> in D2-MSNs produces a similar effect through dopamine D2 receptor. Administration of GlcCer or the PPARγ agonist pioglitazone reverses stress-induced depression-like behaviors, combined GlcCer and pioglitazone exerts additive antidepressant effects. <b>Conclusions:</b> These findings demonstrate a pivotal role for GlcCer-PPARγ signaling in D2-MSNs in depression, highlighting the therapeutic potential of targeting PPARγ activity in depression.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"2845-2865"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775823/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145934959","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}
Feifei Sun, Yuchuan Yan, Deqing Sun, Shijia Liu, Zhaoru Dong, Guoqiang Pan, Lin Zhang, Xianhao Shao, Yuliang Xu, Ying Qu, Tao Li
Rationale: Hormonal therapy is fundamental to prostate cancer (PCa) management; however, its long-term efficacy is compromised by enzalutamide resistance (ENZR), which is fuelled by prostate cancer stem-like cells (PCaSCs) and an immunosuppressive microenvironment. Methods: A CD44-targeted nanoactivator (EC@HNA) was engineered to co-deliver ENZ and siCAMK1D. Its physicochemical properties, cellular uptake and gene-silencing efficiency were characterized in vitro. Functional and mechanistic assays were used to assess PCaSCs expansion, cytokine modulation, immune cell dynamics, and CREB-dependent regulation of stemness genes. Therapeutic efficacy and safety were validated in ENZR cell cultures, murine tumor models, and patient-derived organoids. Results: EC@HNA efficiently delivered siCAMK1D and achieved potent CAMK1D silencing, thereby significantly suppressing the expansion and self-renewal of PCaSCs. This treatment downregulated the immunosuppressive cytokines IL-10 and TGF-β, decreased regulatory T cell (Treg) infiltration, promoted M1-like polarization of tumor-associated macrophages, and enhanced CD8⁺ T cell infiltration and cytotoxicity in ENZR prostate tumors, thereby reprogramming the tumor immune microenvironment. Mechanistically, EC@HNA suppressed CREB phosphorylation at Ser133, which transcriptionally repressed key stemness regulators, including CD44, CD133, and NR4A1, thereby attenuating tumor stemness and immune evasion. These effects have been validated using in vitro cell models, ENZR xenografts, and patient-derived organoids. Collectively, EC@HNA dismantled the stemness-immunity axis sustaining ENZR and restored robust anti-tumor immunity with minimal systemic toxicity. Conclusions: Overall, the CD44-targeted EC@HNA nanoplatform disrupted stemness programs and restored tumor-immune surveillance, representing a promising strategy to reverse ENZR and potentiate immunotherapy in clinical ENZR PCa patients.
{"title":"Targeting CAMK1D-engineered nanoactivator suppresses cancer stem cell maintenance and immune evasion in enzalutamide-resistant prostate cancer.","authors":"Feifei Sun, Yuchuan Yan, Deqing Sun, Shijia Liu, Zhaoru Dong, Guoqiang Pan, Lin Zhang, Xianhao Shao, Yuliang Xu, Ying Qu, Tao Li","doi":"10.7150/thno.120826","DOIUrl":"10.7150/thno.120826","url":null,"abstract":"<p><p><b>Rationale:</b> Hormonal therapy is fundamental to prostate cancer (PCa) management; however, its long-term efficacy is compromised by enzalutamide resistance (ENZR), which is fuelled by prostate cancer stem-like cells (PCaSCs) and an immunosuppressive microenvironment. <b>Methods:</b> A CD44-targeted nanoactivator (EC@HNA) was engineered to co-deliver ENZ and siCAMK1D. Its physicochemical properties, cellular uptake and gene-silencing efficiency were characterized <i>in vitro</i>. Functional and mechanistic assays were used to assess PCaSCs expansion, cytokine modulation, immune cell dynamics, and CREB-dependent regulation of stemness genes. Therapeutic efficacy and safety were validated in ENZR cell cultures, murine tumor models, and patient-derived organoids. <b>Results:</b> EC@HNA efficiently delivered siCAMK1D and achieved potent CAMK1D silencing, thereby significantly suppressing the expansion and self-renewal of PCaSCs. This treatment downregulated the immunosuppressive cytokines IL-10 and TGF-β, decreased regulatory T cell (Treg) infiltration, promoted M1-like polarization of tumor-associated macrophages, and enhanced CD8⁺ T cell infiltration and cytotoxicity in ENZR prostate tumors, thereby reprogramming the tumor immune microenvironment. Mechanistically, EC@HNA suppressed CREB phosphorylation at Ser133, which transcriptionally repressed key stemness regulators, including CD44, CD133, and NR4A1, thereby attenuating tumor stemness and immune evasion. These effects have been validated using <i>in vitro</i> cell models, ENZR xenografts, and patient-derived organoids. Collectively, EC@HNA dismantled the stemness-immunity axis sustaining ENZR and restored robust anti-tumor immunity with minimal systemic toxicity. <b>Conclusions:</b> Overall, the CD44-targeted EC@HNA nanoplatform disrupted stemness programs and restored tumor-immune surveillance, representing a promising strategy to reverse ENZR and potentiate immunotherapy in clinical ENZR PCa patients.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 6","pages":"2918-2935"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12775825/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935008","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}
Ying Bai, Bing Han, Yi Zhang, Liying Wen, Chang Liu, Yu Wang, Jiafang Cui, Bingjing Zheng, Ningbo Cai, Lian Xu, Ling Shen, Yuan Zhang, Honghong Yao
Rationale: Aberrant circular RNA (circRNA) expression is implicated in various diseases, but the regulatory mechanisms remain poorly understood. Our previous work identified circSCMH1 as a brain repair-associated circRNA, prompting investigation into its biogenesis regulation.
Methods: We combined computational analysis of RNA-binding protein (RBP) binding sites in flanking intronic regions with transcriptomic sequencing to identify potential circSCMH1 regulators. Molecular biology experiments including RNA immunoprecipitation and functional assays were performed to validate the interaction between candidate RBPs and circSCMH1 precursor sequences.
Results: Polypyrimidine tract binding protein 1 (PTBP1) was identified as a key regulator binding specifically to the 800-882 segment at the 3' end of circSCMH1's flanking intron. This binding event inhibited back-splicing and reduces circSCMH1 production. Functional studies demonstrated that PTBP1-mediated suppression of circSCMH1 exacerbates post-stroke brain injury.
Conclusions: Our study reveals a novel molecular mechanism whereby PTBP1 regulates circSCMH1 biogenesis through suppression of back-splicing. These findings advance understanding of circRNA regulatory networks and suggest potential therapeutic targets for stroke recovery.
{"title":"PTBP1-mediated inhibition of circular RNA SCMH1 biogenesis impairs brain recovery after ischemic stroke.","authors":"Ying Bai, Bing Han, Yi Zhang, Liying Wen, Chang Liu, Yu Wang, Jiafang Cui, Bingjing Zheng, Ningbo Cai, Lian Xu, Ling Shen, Yuan Zhang, Honghong Yao","doi":"10.7150/thno.114179","DOIUrl":"10.7150/thno.114179","url":null,"abstract":"<p><strong>Rationale: </strong>Aberrant circular RNA (circRNA) expression is implicated in various diseases, but the regulatory mechanisms remain poorly understood. Our previous work identified circSCMH1 as a brain repair-associated circRNA, prompting investigation into its biogenesis regulation.</p><p><strong>Methods: </strong>We combined computational analysis of RNA-binding protein (RBP) binding sites in flanking intronic regions with transcriptomic sequencing to identify potential circSCMH1 regulators. Molecular biology experiments including RNA immunoprecipitation and functional assays were performed to validate the interaction between candidate RBPs and circSCMH1 precursor sequences.</p><p><strong>Results: </strong>Polypyrimidine tract binding protein 1 (PTBP1) was identified as a key regulator binding specifically to the 800-882 segment at the 3' end of circSCMH1's flanking intron. This binding event inhibited back-splicing and reduces circSCMH1 production. Functional studies demonstrated that PTBP1-mediated suppression of circSCMH1 exacerbates post-stroke brain injury.</p><p><strong>Conclusions: </strong>Our study reveals a novel molecular mechanism whereby PTBP1 regulates circSCMH1 biogenesis through suppression of back-splicing. These findings advance understanding of circRNA regulatory networks and suggest potential therapeutic targets for stroke recovery.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 1","pages":"239-253"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12665102/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145655642","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: Spinal cord injury (SCI) leads to permanent sensory and motor function loss, characterized by inflammation and neuronal loss. A promising therapeutic strategy involves delivering anti-inflammatory and neuroregenerative agents tailored to these phases. Methods: GelMA-AFN hydrogel microspheres were prepared by a UV-crosslinked microfluidic chip. Immunofluorescence was performed to assess the effect of GelMA-AFN on apoptosis, axonal growth in dorsal root ganglion (DRG) neurons. Immunohistochemistry, flow cytometry, electrophysiology, RNA-seq, and behavioral testing were used to evaluate histological and functional recovery in a rat SCI model. Results: In this study, we developed GelMA-AFN with a dual-layer structure and an mean diameter of 50 µm. The outer layer, containing low-concentration gelatin methacryloyl (GelMA, 5%) and annexin A1 (ANXA1), provided sustained ANXA1 released for up to 7 days, while the inner layer, with high-concentration GelMA (10%), nanoclay, fibronectin (FN), and nerve growth factor (NGF), releases FN and NGF over 6 weeks. In vitro, GelMA-AFN inhibits neuronal apoptosis, promoted axonal growth, and enhances survival under oxidative stress. In vivo, it reduced early inflammation by limiting neutrophil recruitment and promoting macrophage M2 polarization. Eight weeks post-SCI in a rat model, GelMA-AFN enhanced axonal extension, myelin regeneration, beneficial ECM deposition, and reduced glial scar formation, leading to significant neural electrical signal conduction and motor function recovery. mRNA-seq analysis confirmed GelMA-AFN upregulates genes associated with anti-inflammatory responses and axonal extension while downregulating pro-inflammatory genes. Conclusion: These results suggest GelMA-AFN as a promising therapeutic approach for SCI by providing spatiotemporal delivery aligned with the injury's dynamic stages.
{"title":"Biomimetic core-shell GelMA microspheres co-delivering ANXA1, NGF, and fibronectin enable phase-matched immunomodulation and neurorepair after spinal cord injury.","authors":"Youjun Liu, Chunping Hu, Siyuan He, Renfeng Liu, Yuqi Zhao, Yuhao Wang, Hailiang Xu, Hui Li, Yanming Ma, Botao Lu, Yixiang Ai, Cheng Ju, Weidong Wu, Yifan Wang, Dageng Huang, Dingjun Hao, Zhiyuan Wang, Baorong He, Lei Zhu","doi":"10.7150/thno.120426","DOIUrl":"10.7150/thno.120426","url":null,"abstract":"<p><p><b>Background:</b> Spinal cord injury (SCI) leads to permanent sensory and motor function loss, characterized by inflammation and neuronal loss. A promising therapeutic strategy involves delivering anti-inflammatory and neuroregenerative agents tailored to these phases. <b>Methods:</b> GelMA-AFN hydrogel microspheres were prepared by a UV-crosslinked microfluidic chip. Immunofluorescence was performed to assess the effect of GelMA-AFN on apoptosis, axonal growth in dorsal root ganglion (DRG) neurons. Immunohistochemistry, flow cytometry, electrophysiology, RNA-seq, and behavioral testing were used to evaluate histological and functional recovery in a rat SCI model. <b>Results:</b> In this study, we developed GelMA-AFN with a dual-layer structure and an mean diameter of 50 µm. The outer layer, containing low-concentration gelatin methacryloyl (GelMA, 5%) and annexin A1 (ANXA1), provided sustained ANXA1 released for up to 7 days, while the inner layer, with high-concentration GelMA (10%), nanoclay, fibronectin (FN), and nerve growth factor (NGF), releases FN and NGF over 6 weeks. <i>In vitro</i>, GelMA-AFN inhibits neuronal apoptosis, promoted axonal growth, and enhances survival under oxidative stress. <i>In vivo</i>, it reduced early inflammation by limiting neutrophil recruitment and promoting macrophage M2 polarization. Eight weeks post-SCI in a rat model, GelMA-AFN enhanced axonal extension, myelin regeneration, beneficial ECM deposition, and reduced glial scar formation, leading to significant neural electrical signal conduction and motor function recovery. mRNA-seq analysis confirmed GelMA-AFN upregulates genes associated with anti-inflammatory responses and axonal extension while downregulating pro-inflammatory genes. <b>Conclusion:</b> These results suggest GelMA-AFN as a promising therapeutic approach for SCI by providing spatiotemporal delivery aligned with the injury's dynamic stages.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 1","pages":"483-515"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12665142/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145655749","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}
Boron neutron-capture therapy (BNCT) is a highly precise, cell-level cancer radiotherapy. It exploits the neutron-capture reaction that occurs when low-energy thermal neutrons are absorbed by a boron-10 atom, triggering a nuclear fission reaction that releases high-energy particles to selectively kill cancer cells. BNCT is at the forefront of cancer treatment. Presently, only sodium mercaptoundecahydro-closo-dodecaborate and boron borylphenylalanine (BPA) have been approved as boron drugs for clinical trials by the Food and Drug Administration. However, these drugs still suffer from shortcomings, such as poor targeting, low concentration in cancer cells, a short residence time, and low overall applicability. Conversely, boron clusters are three-dimensional polyhedral structures composed of carbon, boron, and hydrogen atoms. Owing to their excellent stability and unique three-dimensional shape, they are ideal candidates for next-generation boron drugs. These unique features make boron clusters an ideal model for correlating macroscopic properties with the microstructures of substances, providing a valuable framework for the rational design of next-generation boron drugs. Thus, from an interdisciplinary perspective, this review summarizes new strategies for constructing boron clusters, including multi-level structures. We describe key chemical strategies for their functionalization for clinical applications, reveal the multi-scenario applications of their line-functionalized derivatives, and highlight their cross-disciplinary value in precision synthesis, biomedicine, and advanced materials, all with a focus on elucidating the structure-function relationship in boron clusters. Additionally, we explored the latest advancements in the visual evaluation of BNCT, its anticancer mechanism, and exclusive neutron accelerator devices. In summary, the development of novel boron drugs based on functional boron clusters is a prerequisite to resolving the key technical issues in the research and development of new BNCT agents. This review provides insights into the design of new BNCT drugs, as well as related supporting equipment and treatment options, from the perspectives of medicinal chemistry and clinical applications.
{"title":"State-of-the-art boron clusters for boron neutron-capture therapy.","authors":"Weiyao Wang, Enze Zhang, Jiaojiao Shan, Min Zhang, Renwei Cai, Runze Li, Lulian Pang, Baosheng Li, Dejin Zang","doi":"10.7150/thno.123376","DOIUrl":"10.7150/thno.123376","url":null,"abstract":"<p><p>Boron neutron-capture therapy (BNCT) is a highly precise, cell-level cancer radiotherapy. It exploits the neutron-capture reaction that occurs when low-energy thermal neutrons are absorbed by a boron-10 atom, triggering a nuclear fission reaction that releases high-energy particles to selectively kill cancer cells. BNCT is at the forefront of cancer treatment. Presently, only sodium mercaptoundecahydro-closo-dodecaborate and boron borylphenylalanine (BPA) have been approved as boron drugs for clinical trials by the Food and Drug Administration. However, these drugs still suffer from shortcomings, such as poor targeting, low concentration in cancer cells, a short residence time, and low overall applicability. Conversely, boron clusters are three-dimensional polyhedral structures composed of carbon, boron, and hydrogen atoms. Owing to their excellent stability and unique three-dimensional shape, they are ideal candidates for next-generation boron drugs. These unique features make boron clusters an ideal model for correlating macroscopic properties with the microstructures of substances, providing a valuable framework for the rational design of next-generation boron drugs. Thus, from an interdisciplinary perspective, this review summarizes new strategies for constructing boron clusters, including multi-level structures. We describe key chemical strategies for their functionalization for clinical applications, reveal the multi-scenario applications of their line-functionalized derivatives, and highlight their cross-disciplinary value in precision synthesis, biomedicine, and advanced materials, all with a focus on elucidating the structure-function relationship in boron clusters. Additionally, we explored the latest advancements in the visual evaluation of BNCT, its anticancer mechanism, and exclusive neutron accelerator devices. In summary, the development of novel boron drugs based on functional boron clusters is a prerequisite to resolving the key technical issues in the research and development of new BNCT agents. This review provides insights into the design of new BNCT drugs, as well as related supporting equipment and treatment options, from the perspectives of medicinal chemistry and clinical applications.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 1","pages":"417-464"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12665126/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145655585","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}
Rui Ye, Yupei Zhang, Wan Xu, Li Lai, Zhongwei Zhang, Yan Chen, Shugang Qin
Developing therapies for complex brain diseases faces significant challenges due to biological complexity and the stringent blood-brain barrier. While nanomedicine holds promise, traditional R&D paradigms suffer from inefficiency. This review introduces an intelligent theranostic paradigm that integrates high-fidelity brain organoid models, high-throughput screening (HTS/HCS), and Artificial Intelligence (AI). In this closed-loop workflow, organoid platforms serve a diagnostic role, generating predictive data on nanomedicine performance. AI then provides therapeutic guidance by processing this data to drive rational drug design, synthesis, and interaction prediction. This AI-driven convergence is poised to significantly accelerate the development of precisely targeted and individualized nanomedicines, offering new hope for breakthroughs in treating brain diseases.
{"title":"AI and organoid platforms for brain-targeted theranostics.","authors":"Rui Ye, Yupei Zhang, Wan Xu, Li Lai, Zhongwei Zhang, Yan Chen, Shugang Qin","doi":"10.7150/thno.123243","DOIUrl":"10.7150/thno.123243","url":null,"abstract":"<p><p>Developing therapies for complex brain diseases faces significant challenges due to biological complexity and the stringent blood-brain barrier. While nanomedicine holds promise, traditional R&D paradigms suffer from inefficiency. This review introduces an intelligent theranostic paradigm that integrates high-fidelity brain organoid models, high-throughput screening (HTS/HCS), and Artificial Intelligence (AI). In this closed-loop workflow, organoid platforms serve a diagnostic role, generating predictive data on nanomedicine performance. AI then provides therapeutic guidance by processing this data to drive rational drug design, synthesis, and interaction prediction. This AI-driven convergence is poised to significantly accelerate the development of precisely targeted and individualized nanomedicines, offering new hope for breakthroughs in treating brain diseases.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"876-897"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674940/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678695","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}
Shaojun Liu, Oxana Semyachkina-Glushkovskaya, Tingting Yu, Egor Ilukov, Edik Rafailov, Sergey Sokolovski, Jürgen Kurths, Dan Zhu
Pharmacological treatment of brain diseases is hampered by the blood-brain barrier that prevents the vast majority of drugs from entering the brain. For this reason, the pharmaceutical industry is reluctant to invest in the development of new neurotropic drugs. Even if effective pharmacological strategies for the treatment of brain diseases will be found, it will take 10-15 years between the emergence of an idea and the introduction of a drug to the market. This creates priority for the development of neuro-lymphaphotonics based on the development of promising non-pharmacological strategies for managing functions of the meningeal lymphatic vessels (MLVs). MLVs play a crucial role in the removal of toxins and metabolites from brain as well as in regulation of brain homeostasis and its immunity. Since MLVs are located on the brain surface, light penetrating the skull easily reaches MLVs and affects their functions. Therefore, MLVs are an ideal target for photobiomodulation (PBM). The pioneering studies have shown that PBM of MLVs is a promising strategy for the treatment of a wide range of neuropathology, including Alzheimer's or age-related brain diseases, brain tumor, intracranial hemorrhage, brain damages caused by diabetes. It has recently been discovered that sleep enhances the therapeutic effects of PBM and is a "therapeutic window" in overcoming the limitations of PBM in the elderly. Considering that the PBM technologies are non-invasive and safe with commercially viable possibilities (portability and low cost), neuro-lymphaphotonics open up promising prospects for the development of future technologies for the effective therapy of brain diseases.
{"title":"Neuro-lymphaphotonics opens new horizons of the future technologies for the therapy of brain diseases.","authors":"Shaojun Liu, Oxana Semyachkina-Glushkovskaya, Tingting Yu, Egor Ilukov, Edik Rafailov, Sergey Sokolovski, Jürgen Kurths, Dan Zhu","doi":"10.7150/thno.120374","DOIUrl":"10.7150/thno.120374","url":null,"abstract":"<p><p>Pharmacological treatment of brain diseases is hampered by the blood-brain barrier that prevents the vast majority of drugs from entering the brain. For this reason, the pharmaceutical industry is reluctant to invest in the development of new neurotropic drugs. Even if effective pharmacological strategies for the treatment of brain diseases will be found, it will take 10-15 years between the emergence of an idea and the introduction of a drug to the market. This creates priority for the development of neuro-lymphaphotonics based on the development of promising non-pharmacological strategies for managing functions of the meningeal lymphatic vessels (MLVs). MLVs play a crucial role in the removal of toxins and metabolites from brain as well as in regulation of brain homeostasis and its immunity. Since MLVs are located on the brain surface, light penetrating the skull easily reaches MLVs and affects their functions. Therefore, MLVs are an ideal target for photobiomodulation (PBM). The pioneering studies have shown that PBM of MLVs is a promising strategy for the treatment of a wide range of neuropathology, including Alzheimer's or age-related brain diseases, brain tumor, intracranial hemorrhage, brain damages caused by diabetes. It has recently been discovered that sleep enhances the therapeutic effects of PBM and is a \"therapeutic window\" in overcoming the limitations of PBM in the elderly. Considering that the PBM technologies are non-invasive and safe with commercially viable possibilities (portability and low cost), neuro-lymphaphotonics open up promising prospects for the development of future technologies for the effective therapy of brain diseases.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"776-793"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674938/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678945","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}
Guangyin Pu, Yayue Song, Qiushi Li, Jinjie Duan, Guangyan Wang, Wenjing Xiu, Jingwen Xu, Xiaoyu Zhao, Wenhui Dong, Tingting Lan, Rong Ai, Jingyi Zhang, Weiyan Sun, Deling Kong, Yi Zhu, Xu Zhang, Yang Liu, Chunjiong Wang
Rationale: Metabolic remodeling occurs during partial hepatectomy (PHx)-induced liver regeneration. Phospholipid remodeling during this process and its subsequent impact on liver regeneration remain unknown. The remnant liver's ability to defend against injury is also essential for normal liver regeneration, although the underlying mechanisms remain unclear. Methods: Phospholipidomics was performed to describe phospholipid remodeling after 70% PHx. Phosphate cytidylyltransferase 2, ethanolamine (PCYT2) was overexpressed in hepatocytes using adeno-associated virus under the thyroxine-binding globulin promoter. An ex vivo liver perfusion system was used to regulate portal pressure. GalNAc-conjugated PEG-PCL nano-particles (NPs) were developed to deliver the PCYT2 inhibitor, meclizine. Results: We found a significant decrease in a series of phosphatidylethanolamine (PE) levels at 1 day after 70% PHx. PCYT2, an enzyme for PE synthesis, was downregulated by PHx. Higher portal pressure-induced shear stress is an early event after PHx. As a target gene of hepatocyte nuclear factor 4α, PCYT2 levels were decreased by higher portal pressure. Hepatocyte-specific PCYT2 overexpression aggravated liver damage after PHx by increasing reactive oxygen species levels, lipid peroxidation, and mitochondrial fragmentation. We observed higher hepatic PCYT2 levels in middle-aged mice than in young mice. PCYT2 inhibition by meclizine facilitates liver regeneration in middle-aged mice. Meclizine is also a blocker of the histamine H1 receptor, a membrane receptor. Therefore, we used NPs to deliver meclizine into cells to better target PCYT2 and prevent potential side effects. NP-meclizine improved liver regeneration in middle-aged mice, demonstrating higher therapeutic efficacy than carrier-free meclizine. Conclusions: Decreased PCYT2 levels and PE content due to increased portal pressure protect hepatocytes from PHx-induced injury. Inhibiting PCYT2 with NP-meclizine promoted normal liver regeneration in middle-aged mice.
{"title":"Downregulation of PCYT2 by increased portal pressure safeguards liver regeneration after partial hepatectomy.","authors":"Guangyin Pu, Yayue Song, Qiushi Li, Jinjie Duan, Guangyan Wang, Wenjing Xiu, Jingwen Xu, Xiaoyu Zhao, Wenhui Dong, Tingting Lan, Rong Ai, Jingyi Zhang, Weiyan Sun, Deling Kong, Yi Zhu, Xu Zhang, Yang Liu, Chunjiong Wang","doi":"10.7150/thno.118755","DOIUrl":"10.7150/thno.118755","url":null,"abstract":"<p><p><b>Rationale:</b> Metabolic remodeling occurs during partial hepatectomy (PHx)-induced liver regeneration. Phospholipid remodeling during this process and its subsequent impact on liver regeneration remain unknown. The remnant liver's ability to defend against injury is also essential for normal liver regeneration, although the underlying mechanisms remain unclear. <b>Methods:</b> Phospholipidomics was performed to describe phospholipid remodeling after 70% PHx. Phosphate cytidylyltransferase 2, ethanolamine (PCYT2) was overexpressed in hepatocytes using adeno-associated virus under the thyroxine-binding globulin promoter. An <i>ex vivo</i> liver perfusion system was used to regulate portal pressure. GalNAc-conjugated PEG-PCL nano-particles (NPs) were developed to deliver the PCYT2 inhibitor, meclizine. <b>Results:</b> We found a significant decrease in a series of phosphatidylethanolamine (PE) levels at 1 day after 70% PHx. PCYT2, an enzyme for PE synthesis, was downregulated by PHx. Higher portal pressure-induced shear stress is an early event after PHx. As a target gene of hepatocyte nuclear factor 4α, PCYT2 levels were decreased by higher portal pressure. Hepatocyte-specific PCYT2 overexpression aggravated liver damage after PHx by increasing reactive oxygen species levels, lipid peroxidation, and mitochondrial fragmentation. We observed higher hepatic PCYT2 levels in middle-aged mice than in young mice. PCYT2 inhibition by meclizine facilitates liver regeneration in middle-aged mice. Meclizine is also a blocker of the histamine H1 receptor, a membrane receptor. Therefore, we used NPs to deliver meclizine into cells to better target PCYT2 and prevent potential side effects. NP-meclizine improved liver regeneration in middle-aged mice, demonstrating higher therapeutic efficacy than carrier-free meclizine. <b>Conclusions:</b> Decreased PCYT2 levels and PE content due to increased portal pressure protect hepatocytes from PHx-induced injury. Inhibiting PCYT2 with NP-meclizine promoted normal liver regeneration in middle-aged mice.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"810-829"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674937/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678797","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}
Zhenjia Che, Xiao Sheng, Yanglin Wu, Binghao Lin, Kaihang Song, Qiyun Chen, Aopan Chen, Lingxuan Deng, Jing Chen, Ming Cai
Rationale: Repairing bone defects in osteoporotic patients presents a significant clinical challenge due to inadequate osseointegration, persistent inflammation, and elevated oxidative stress. To overcome these barriers, this study proposes the development of a functionalized 3D-printed titanium alloy porous scaffold capable of sequentially releasing therapeutic agents to modulate the immune environment and enhance bone regeneration. Methods: A thermosensitive collagen hydrogel was integrated with a zeolitic imidazolate framework (ZIF-8) to construct a dual-release platform capable of delivering the immunomodulator 4-octyl itaconate (4-OI) and the osteogenic factor bone morphogenetic protein-9 (BMP-9) in a temporally controlled manner. The hydrogel facilitated early-phase release of 4-OI to inhibit M1 macrophage polarization and mitigate oxidative stress, while ZIF-8 enabled sustained BMP-9 release to induce osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Comprehensive in vitro assays and an osteoporotic rat model were employed to evaluate the scaffold's immunomodulatory properties, osteogenic capacity, and osseointegration performance. Results: The scaffold inhibited pro-inflammatory cytokine expression, attenuated osteoclast activity, and enhanced osteogenic marker levels in vitro. In vivo analysis revealed enhanced bone-implant interface integration and significantly accelerated bone regeneration in osteoporotic defects. Transcriptome analysis revealed suppression of NF-κB and TGF-β signaling, confirming the scaffold's combined immunomodulatory and osteoinductive effects. Conclusions: This ZIF-functionalized hydrogel scaffold with sequential release capability offers a potential strategy for clinical translation in osteoporotic bone defect repair. By orchestrating local immune modulation and promoting sustained osteogenesis, the system offers a clinically relevant approach to enhance osseointegration and facilitate long-term bone repair in osteoporotic conditions.
{"title":"Sequential release of bioactive factors from functionalized metal-organic framework hydrogel enhances interfacial osseointegration of 3D-printed titanium alloy porous scaffolds.","authors":"Zhenjia Che, Xiao Sheng, Yanglin Wu, Binghao Lin, Kaihang Song, Qiyun Chen, Aopan Chen, Lingxuan Deng, Jing Chen, Ming Cai","doi":"10.7150/thno.120711","DOIUrl":"10.7150/thno.120711","url":null,"abstract":"<p><p><b>Rationale:</b> Repairing bone defects in osteoporotic patients presents a significant clinical challenge due to inadequate osseointegration, persistent inflammation, and elevated oxidative stress. To overcome these barriers, this study proposes the development of a functionalized 3D-printed titanium alloy porous scaffold capable of sequentially releasing therapeutic agents to modulate the immune environment and enhance bone regeneration. <b>Methods:</b> A thermosensitive collagen hydrogel was integrated with a zeolitic imidazolate framework (ZIF-8) to construct a dual-release platform capable of delivering the immunomodulator 4-octyl itaconate (4-OI) and the osteogenic factor bone morphogenetic protein-9 (BMP-9) in a temporally controlled manner. The hydrogel facilitated early-phase release of 4-OI to inhibit M1 macrophage polarization and mitigate oxidative stress, while ZIF-8 enabled sustained BMP-9 release to induce osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Comprehensive in vitro assays and an osteoporotic rat model were employed to evaluate the scaffold's immunomodulatory properties, osteogenic capacity, and osseointegration performance. <b>Results:</b> The scaffold inhibited pro-inflammatory cytokine expression, attenuated osteoclast activity, and enhanced osteogenic marker levels in vitro. In vivo analysis revealed enhanced bone-implant interface integration and significantly accelerated bone regeneration in osteoporotic defects. Transcriptome analysis revealed suppression of NF-κB and TGF-β signaling, confirming the scaffold's combined immunomodulatory and osteoinductive effects. <b>Conclusions:</b> This ZIF-functionalized hydrogel scaffold with sequential release capability offers a potential strategy for clinical translation in osteoporotic bone defect repair. By orchestrating local immune modulation and promoting sustained osteogenesis, the system offers a clinically relevant approach to enhance osseointegration and facilitate long-term bone repair in osteoporotic conditions.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":"16 2","pages":"852-875"},"PeriodicalIF":13.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12674936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145678875","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}