2025-05-20 2025, Volume 6 Issue 5

  • Select all
  • ORIGINAL ARTICLE
    Houshi Xu , Zhen Fan , Shan Jiang , Maoyuan Sun , Huihui Chai , Ruize Zhu , Xiaoyu Liu , Yue Wang , Jiawen Chen , Junji Wei , Ying Mao , Zhifeng Shi

    Glioma subtyping is crucial for treatment decisions, but traditional approaches often fail to capture tumor heterogeneity. This study proposes a novel framework integrating multiplex immunohistochemistry (mIHC) and machine learning for glioma subtyping and prognosis prediction. 185 patient samples from the Huashan hospital cohort were stained using a multi-label mIHC panel and analyzed with an AI-based auto-scanning system to calculate cell ratios and determine the proportion of positive tumor cells for various markers. Patients were divided into two cohorts (training: N = 111, testing: N = 74), and a machine learning model was then developed and validated for subtype classification and prognosis prediction. The framework identified two distinct glioma subtypes with significant differences in prognosis, clinical characteristics, and molecular profiles. The high-risk subtype, associated with older age, poorer outcomes, astrocytoma/glioblastoma, higher tumor grades, elevated mesenchymal scores, and an inhibitory immune microenvironment, exhibited IDH wild-type, 1p19q non-codeletion, and MGMT promoter unmethylation, suggesting chemotherapy resistance. Conversely, the low-risk subtype, characterized by younger age, better prognosis, astrocytoma/oligodendroglioma, lower tumor grades, and favorable molecular profiles (IDH mutation, 1p19q codeletion, MGMT promoter methylation), indicated chemotherapy sensitivity. The mIHC-based framework enables rapid glioma classification, facilitating tailored treatment strategies and accurate prognosis prediction, potentially improving patient management and outcomes.

  • ORIGINAL ARTICLE
    Tao Chen , Yang Zhang , Liting Ding , Chenlu Xiong , Chao Mei , Sisi Wei , Ming Jiang , Yingjie Huang , Jianrong Chen , Tao Xie , Qing Zhu , Qi Zhang , Xuan Huang , Shibiao Chen , Yong Li

    Acute kidney injury (AKI) is a prevalent and serious clinical disease with a high incidence rate and significant health burden. The limited understanding of the complex pathological mechanisms has hindered the development of efficacious therapeutics. Tripartite motif containing 65 (TRIM65) has recently been identified as a key regulator of acute inflammation. However, its role in AKI remains unclear. The present study observed that TRIM65 expression was upregulated in AKI. Moreover, the knockout of the Trim65 gene in mice exhibited a substantial protective impact against rhabdomyolysis, ischemia-reperfusion (I/R), and cisplatin-induced AKI. Mechanistically, TRIM65 directly binds and mediates K48/K63-linked polyubiquitination modifications of voltage-dependent anion channel 1 (VDAC1) at its K161 and K200 amino acid sites. TRIM65 plays a role in maintaining the stability of VDAC1 and preventing its degradation by the autophagy pathway. TRIM65 deficiency attenuates mitochondrial dysfunction in renal tubular epithelial cells during AKI. Conversely, the overexpression of VDAC1 in renal tissues has been demonstrated to negate the protective effect of TRIM65 deficiency on AKI. These findings suggest that TRIM65 may play a role regulating of AKI through the targeting of VDAC1-dependent mitochondrial function, offering potential avenues for the development of new drug targets and strategies for the treatment of AKI.

  • ORIGINAL ARTICLE
    Maosen Xu , Li Xu , Tao Zhang , Xue Li , Ziqi Zhang , Ruolan Xia , Ning Jiang , Li Yang , Xiawei Wei

    Lung cancer continues to be the primary cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) being the predominant type. Dysregulation of protein translation that participates in cell proliferation is an important factor to define oncogenic processes and cancer development. The eukaryotic initiation factor 4E (eIF4E) regulates ribosomal translation of proteins from mRNA, and the mitogen-activated protein kinase interacting kinases (MNKs) is reported to be the only kinases that can phosphorylate eIF4E. Substantial previous work has proven that the MNK–eIF4E axis is usually dysregulated in many cancer types. Moreover, abnormal angiogenesis is essential for tumorigenesis and cancer progression, and vascular endothelial growth factors (VEGF) together with their receptors play multiple crucial roles in angiogenesis, especially VEGFR2. In this study, we report a novel dual MNK/VEGFR2 inhibitor named JDB153 and investigate its antitumor effects in NSCLC. JDB153 can effectively inhibit the phosphorylation of eIF4E and VEGFR2, suppress proliferation, migration and invasion, promote apoptosis, and induce cycle arrest of lung cancer cells. Importantly, JDB153 exhibits antitumor activity and synergizes with anti-PD-1 therapy and cisplatin with reliable safety. Our findings reveal the potential value of JDB153 in lung cancer as monotherapy or in combination with immunotherapy and chemotherapy, with the hope to provide a novel combinational strategy for NSCLC treatment clinically.

  • ORIGINAL ARTICLE
    Xiaoxu Wang , Lu Dai , Na Wu , Donghui Wu , Xinyuan Wang , Xia Meng , Qilei Zhang , Jing Lu , Xiaoxin Yan , Jing Zhang , Baian Chen

    There are no effective curative treatments for Alzheimer's disease (AD), the most prevalent form of dementia. Amyloid-beta (Aβ) oligomers are considered key neurotoxic molecules that trigger AD. Recent studies have shown that direct antibody targeting of Aβ oligomers is beneficial for early AD patients; however, serious side effects (e.g., brain hemorrhage, edema, and shrinkage) persist. Considering that Aβ oligomers readily bind to other proteins, contributing to neurotoxicity and AD onset, those proteins could represent alternative therapeutic targets. However, proteins that bind to Aβ oligomers in the brains of AD patients have not yet been identified. In this study, we identified four proteins (DDX6, DSP, JUP, and HRNR) that bind to Aβ oligomers derived from the brains of AD patients. Intriguingly, among these four proteins, only the blockade of DEAD-box helicase 6 (DDX6) in human-derived Aβ oligomers attenuated their neurotoxicity both in vitro and in vivo. Mechanistic analysis revealed that DDX6 promotes the formation of Aβ oligomers, likely due to DDX6 bind to Aβ oligomers at four distinct sites. These findings suggest that DDX6 could serve as a potential therapeutic target to reduce the neurotoxicity of Aβ oligomers in the brain and prevent the progression of AD.

  • ORIGINAL ARTICLE
    Zengyu Zhang , Dewen Ru , Zhuohang Liu , Zimin Guo , Lei Zhu , Yuan Zhang , Min Chu , Yong Wang , Jing Zhao

    Chronic cerebral hypoperfusion (CCH) is a significant factor that accelerates cognitive deterioration, yet the mechanisms of hippocampal microglial activation in this context remain unclear. Using an integrative multiomics approach, we investigated the transcriptional and epigenomic landscape of microglial activation in a mouse model of CCH induced by bilateral common carotid artery stenosis. Behavioral assessments revealed cognitive impairments, while neuropathological analysis confirmed hippocampal damage. Proteomic and transcriptomic profiling uncovered significant upregulation of stress and inflammatory pathways, particularly the interferon (IFN) signaling cascade. Epigenomic analysis identified regions of open chromatin, suggesting active transcriptional regulation driven by the transcription factor (TF) PU.1. ChIP-nexus analysis further confirmed that PU.1 directly modulates the expression of IFN-stimulated genes (ISGs), which are pivotal in regulating microglial activation. Our findings demonstrate that PU.1 serves as a key regulator of the IFN-driven microglial response during CCH, mediated by enhanced chromatin accessibility and transcriptional activation of ISGs. This study highlights the critical role of PU.1 in microglial-mediated neuroinflammation and offers potential therapeutic targets for mitigating hippocampal damage associated with chronic cerebral ischemia.

  • REVIEW
    Ting Zhao , Xiaowei Zhang , Xin Liu , Qifeng Wang , Xichun Hu , Zhiguo Luo

    Cancer of unknown primary (CUP), a set of histologically confirmed metastases that cannot be identified or traced back to its primary despite comprehensive investigations, accounts for 2–5% of all malignancies. CUP is the fourth leading cause of cancer-related deaths worldwide, with a median overall survival (OS) of 3–16 months. CUP has long been challenging to diagnose principally due to the occult properties of primary site. In the current era of molecular diagnostics, advancements in methodologies based on cytology, histology, gene expression profiling (GEP), and genomic and epigenomic analysis have greatly improved the diagnostic accuracy of CUP, surpassing 90%. Our center conducted the world's first phase III trial and demonstrated improved progression-free survival and favorable OS by GEP-guided site-specific treatment of CUP, setting the foundation of site-specific treatment in first-line management for CUP. In this review, we detailed the epidemiology, etiology, pathogenesis, as well as the histologic, genetic, and clinical characteristics of CUP. We also provided an overview of the advancements in the diagnostics and therapeutics of CUP over the past 50 years. Moving forward, we propose optimizing diagnostic modalities and exploring further-line treatment regimens as two focus areas for future studies on CUP.

  • REVIEW
    Weijie Xie , Yuan Li , Xinyue Wang , Elena Blokhina , Evgeny Krupitsky , Marina Vetrova , Ji Hu , Ti-Fei Yuan , Jue Chen , Hua Wang , Xiangfang Chen

    Gamma-aminobutyric acid (GABA) B receptors (GABABRs) that acts slowly and maintains the inhibitory tone are versatile regulators in the complex nervous behaviors and their involvement in various neuropsychiatric disorders, such as anxiety, epilepsy, pain, drug addiction, and Alzheimer's disease. Additional study advances have implied the crucial roles of GABABRs in regulating feeding-related behaviors, yet their therapeutic potential in addressing the neuropsychiatric disorders, binge eating, and feeding-related disorders remains underutilized. This general review summarized the physiological structure and functions of GABABR, explored the regulation in various psychiatric disorders, feeding behaviors, binge eating, and metabolism disorders, and fully discussed the potential of targeting GABABRs and its regulator-binding sites for the treatment of different psychiatric disorders, binge eating and even obesity. While agonists that directly bind to GABABR1 have some negative side effects, positive allosteric modulators (PAMs) that bind to GABABR2 demonstrate excellent therapeutic efficacy and tolerability and have better safety and therapeutic indexes. Moreover, phosphorylation sites of downstream GABABRs regulators may be novel therapeutic targets for psychiatric disorders, binge eating, and obesity. Further studies, clinical trials in particular, will be essential for confirming the therapeutic value of PAMs and other agents targeting the GABABR pathways in a clinical setting.

  • ORIGINAL ARTICLE
    Yong-Qing Dou , Xiao-Yun Zhang , Rui-Juan Guo , Xiao-Fu Huang , Yu Song , Xin-Long Liu , Jie Shi , Fan-Qin Li , Dan-Dan Zhang , Peng Kong , Lei Nie , Han Li , Fan Zhang , Mei Han

    Vascular smooth muscle cell (VSMC) plasticity is crucial for the repair after vascular injury. However, the high plasticity of VSMCs may make them transform into pathogenic phenotypes. Here, we show that VSMCs overexpressing Sirtuin 1 (SIRT1) exhibit a reduced phenotypic plasticity in the context of platelet-derived growth factor (PDGF)-BB treatment. SIRT1 activated Quaking (QKI)–cZFP609 axis is involved in the plasticity regulation in the VSMCs. Mechanically, SIRT1 deacetylates K133 and K134 of QKI and mediates its activation. Activated QKI binds the QKI response elements located in the upstream and downstream of the cZFP609-forming exons in ZFP609 pre-mRNA to mediate cZFP609 production. Furthermore, the acetylation of QKI is increased by inhibiting SIRT1 with the selective and potent inhibitor EX527 or deletion of SIRT1, accompanied with parallel decrease in cZFP609 formation. Final, we identify that cZFP609 directs PDGF receptor (PDGFR)β sorting into endosomal/lysosomal pathway and degradation by bridging PDGFRβ and Rab7, resulted in attenuating Raf–MEK–ERK cascade activation downstream of PDGFRβ signaling. Overexpression of cZFP609 remedies aberrant plasticity and overproliferation of VSMCs, and ameliorates neointimal formation. Together, these results highlight that modulating the QKI–cZFP609 axis may help propel repair without stenosis as a therapeutic strategy in vascular injury.

  • REVIEW
    Yao Shen , Nairui Fan , Shu-xia Ma , Xin Cheng , Xuesong Yang , Guang Wang

    Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut–brain axis, gut–liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.

  • ORIGINAL ARTICLE
    Jacopo Romoli , Paola Chiodelli , Patrizia Bonassi Signoroni , Elsa Vertua , Clarissa Ferrari , Elisabetta Giuzzi , Alice Paini , Elisa Scalvini , Andrea Papait , Francesca Romana Stefani , Antonietta Rosa Silini , Ornella Parolini

    The tumor microenvironment (TME) is the combination of cells and factors that promotes tumor progression, and cancer-associated fibroblasts (CAFs) are a key component within TME. CAF originates from various stromal cells and is activated by factors such as transforming growth factor-beta (TGF-β) secreted by tumor cells, favoring chemoresistance and metastasis. Recent publications have underlined plasticity and heterogeneity and their strong contribution to the reactive stroma within the TME. Our study aimed to replicate the TME's structure by creating a 3D in vitro model of ovarian cancer (OC). By incorporating diverse tumor and stromal cells, we simulated a physiologically relevant environment for studying CAF-like cell behavior within tumor spheroids in a context-dependent manner. CAF-like cells were generated by exposing human dermal fibroblasts to OC cell line conditioned media in the presence or absence of TGF-β. Herein, we found that different stimuli induce the generation of heterogeneous populations of CAF-like cells. Notably, we observed the ability of CAF-like cells to shape the intratumoral architecture and to contribute to functional changes in tumor cell behavior. This study highlights the importance of precise assessment of CAF for potential therapeutic interventions and further provides a reliable model for investigating novel therapeutic targets in OC.

  • ORIGINAL ARTICLE
    Xu Guo , Chao Zhang , Yuzhen Wang , Zhao Li , Yaxin Tan , Dongzhen Zhu , Wei Song , Yi Kong , Jinpeng Du , Yuyan Huang , Liting Liang , Jianjun Li , Mengde Zhang , Linhao Hou , Qinhua Liu , Feng Tian , Bingyang Yu , Yue Kong , Zhenyu Zhou , Xiaobing Fu , Sha Huang

    Sudomotor dysfunction in diabetic patients increases the risk of fissures, infections, and diabetic foot ulcers (DFUs), thereby reducing the quality of life. Despite its clinical importance, the mechanisms underlying this dysfunction remain inadequately elucidated. This study addresses this gap by demonstrating that despite structural integrity, sweat glands (SGs) in diabetic individuals with DFUs, and a murine model of diabetic neuropathy (DN), exhibit functional impairments, as confirmed by histological and functional assays. Integrated transcriptome and proteome analysis revealed significant upregulation of the SG microenvironment in response to hypoxia, highlighting potential underlying pathways involved. In addition, histological staining and tissue clearing techniques provided evidence of impaired neurovascular networks adjacent to SGs. Single-cell RNA sequencing unveiled intricate intercellular communication networks among endothelial cells (ECs), neural cells (NCs), and sweat gland cells (SGCs), emphasizing intricate cellular interactions within the SG microenvironment. Furthermore, an in vitro SGC–NC interaction model (SNIM) was employed to validate the supportive role of NCs in regulating SGC functions, highlighting the neurovascular-SG axis in diabetic pathophysiology. These findings confirm the hypoxia-driven upregulation of the SG microenvironment and underscore the critical role of the neurovascular-SG axis in diabetic pathophysiology, providing insights into potential therapeutic targets for managing diabetic complications and improving patient outcomes.

  • ORIGINAL ARTICLE
    Yicheng Yang , Zhiwei Zeng , Qiaoxi Yang , Huan Wang , Hanwen Zhang , Wenjie Yan , Peizhi Wang , Chuangshi Wang , Zhanhao Su , Pugazhenthan Thangaraju , Sher Zaman Safi , Beilan Yang , Yaoyao Wang , Jingjing Zhou , Zhiyong Zou , Yuan Huang , Songren Shu , Changming Xiong

    Pulmonary arterial hypertension (PAH) poses significant clinical management challenges due to gaps in understanding its global epidemiology. We analyzed PAH-related disability-adjusted life years (DALYs), deaths, and prevalence from 1990 to 2021. Age-period-cohort models and regression analyses assessed temporal trends and projected burdens to 2050. Globally, PAH-related DALYs declined by 6.6%, but increased by 13.9% in high socio-demographic index (SDI) countries. Middle SDI regions reported the highest DALYs in 1990 and 2021. Deaths rose by 48.5% worldwide, with high SDI nations experiencing a 76.6% surge. Age-standardized rates (ASRs) of DALYs and deaths decreased across SDI countries, with high-middle SDI regions showing the steepest declines. Younger age groups, especially males, had a higher proportion of global DALYs in earlier years, but the burden shifted toward older populations over time, with this trend more pronounced in high-SDI countries. Age-period-cohort analysis revealed declining DALYs in younger ages but rising rates in older cohorts. By 2050, deaths and prevalence are projected to rise, disproportionately affecting females. Significant regional disparities in PAH burden persist, necessitating targeted policies, improved healthcare access, and early detection strategies, especially in underserved areas. Addressing these disparities is critical for mitigating PAH’ s global impact.

  • REVIEW
    Peter Illes , Patrizia Rubini , Henning Ulrich , Hai-Yan Yin , Yong Tang

    The gliotransmitter adenosine 5'-triphosphate (ATP) and its enzymatic degradation product adenosine play a major role in orchestrating in the hippocampus cognitive and affective functions via P2 purinoceptors (P2X, P2Y) and P1 adenosine receptors (A1, A2A). Although numerous reviews exist on purinoceptors that modulate these functions, there is an apparent gap relating to the involvement of astrocyte-derived extracellular ATP. Our review focuses on the following issues: An impeded release of ATP from hippocampal astrocytes through vesicular mechanisms or connexin hemichannels and pannexin channels interferes with spatial working memory in rodents. The pharmacological blockade of P2Y1 receptors (P2Y1Rs) reverses the deficits in learning/memory performance in mouse models of familial Alzheimer's disease (AD). Similarly, in mouse models of major depressive disorder (MDD), based on acute or chronic stress-induced development of depressive-like behavior, a reduced exocytotic/channel-mediated ATP release from hippocampal astrocytes results in the deterioration of these behavioral responses. However, on the opposite, the increased stimulation of the microglial/astrocytic P2X7R-channel by ATP causes neuroinflammation and in consequence depressive-like behavior. In conclusion, there is strong evidence for the assumption that gliotransmitter ATP is intimately involved in the pathophysiology of cognitive and affective neuron/astrocyte-based human illnesses opening new diagnostic and therapeutic vistas for AD and MDD.

  • HIGHLIGHT
    Kai Huang , Anqi Zhou , Xiangxiang Zhou
  • ORIGINAL ARTICLE
    Ying Zhang , Feiyin Zhu , Zhe Zhang , Jing Wang , Tianyi Liao , Yu Xi , Defeng Liu , Haitao Zhang , Haocheng Lin , Jiaming Mao , Wenhao Tang , Lianming Zhao , Peng Yuan , Liying Yan , Qiang Liu , Kai Hong , Jie Qiao

    The emergence of coronavirus disease 2019 (COVID-19) has triggered research into its impact on male reproductive health. However, studies exploring the effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on semen quality in infertile men remain limited. Herein, we enrolled 781 male infertile patients who recovered from COVID-19 and analyzed their semen and blood samples collected at different time points. We found that SARS-CoV-2 RNA was undetectable in semen samples. Compared with pre-COVID-19 status, total sperm count, sperm concentration, vitality, motility, and percentage of sperm cells with normal morphology decreased significantly in the first month post-COVID-19. However, these alterations were reversed in the third month. Furthermore, seminal plasma samples exhibited reduced proinflammatory cytokine levels and notable changes in amino acid, nucleic acid, and carbohydrate metabolism by the third month compared with those in the first month. By contrast, no significant alterations in reproductive hormone levels were found. Vitality, progressive motility, and total motility negatively correlated with body temperature when it was above 38°C. In conclusion, semen quality initially decreases post-COVID-19 but reverses after approximately 3 months, with a decline related to inflammatory and fever. These findings may provide guidance to infertile male patients who need assisted reproductive technology.

  • REVIEW
    Sepideh Hassibian , Mahsa Amin , Seyed Mohammad Taghdisi , Elham Sameiyan , Reza Ghaffari , Mona Alibolandi , Mohammad Ramezani , Khalil Abnous , Seyed Mohsen Dehnavi

    Single-stranded DNA or RNA entities referred to as aptamers exhibit a strong affinity and specificity for attaching to specific targets. Owing to their special properties, which include simplicity of synthesis, low immunogenicity, and adaptability in targeting a variety of substances, these synthetic oligonucleotides have garnered a lot of interest. The function of aptamers can be altered by combining them with complementary oligonucleotides “antidotes,” which are antisense to a particular aptamer sequence. Antidotes play an important role in several fields by specifically targeting the corresponding section of the aptamer. Nevertheless, even with their promising capabilities, the creation of antidotes to regulate or inhibit aptamer function continues to be a relatively unexamined field, constraining their secure and efficient application in medical environments. The review explores experimental methodologies for creating antidotes, the systematic design strategies for managing antidotes in aptamer-based therapies, and their therapeutic efficacy in counteracting disease biomarkers. Additionally, it highlights their diagnostic applications in biosensing and imaging, offering a promising alternative to traditional antibodies. It also investigates the progress, latest innovations, and potential medical uses of aptamer–antidote combinations. Its academic value lies in bridging the gap between theoretical design and practical applications, providing researchers and clinicians with a comprehensive resource to advance aptamer-based solutions in medicine and biotechnology.

  • REVIEW
    Lingling Xue , Ran An , Junqi Zhao , Mengdi Qiu , Zhongxia Wang , Haozhen Ren , Decai Yu , Xinhua Zhu

    Hydrogels have emerged as dependable candidates for tissue repair because of their exceptional biocompatibility and tunable mechanical properties. However, conventional hydrogels are vulnerable to damage owing to mechanical stress and environmental factors that compromise their structural integrity and reduce their lifespan. In contrast, self-healing hydrogels with their inherent ability to restore structure and function autonomously offer prolonged efficacy and enhanced appeal. These hydrogels can be engineered into innovative forms including stimulus-responsive, self-degradable, injectable, and drug-loaded variants, thereby enhancing their applicability in wound healing, drug delivery, and tissue engineering. This review summarizes the categories and mechanisms of self-healing hydrogels, along with their biomedical applications, including tissue repair, drug delivery, and biosensing. Tissue repair includes wound healing, bone-related repair, nerve repair, and cardiac repair. Additionally, we explored the challenges that self-healing hydrogels continue to face in tissue repair and presented a forward-looking perspective on their development. Consequently, it is anticipated that self-healing hydrogels will be progressively designed and developed for applications that extend beyond tissue repair to a broader range of biomedical applications.

  • REVIEW
    Tianyou Ma , Tingrui Zhang , Fengze Miao , Jun Liu , Quangang Zhu , Zhongjian Chen , Zongguang Tai , Zhigao He

    Alopecia areata (AA) is a complex, chronic inflammatory skin disorder characterized by unpredictable, nonscarring hair loss, affecting millions worldwide. Its pathogenesis remains poorly understood, driven by intricate interactions among immune dysregulation, genetic predisposition, and environmental triggers. Despite significant advances in identifying these contributing factors, substantial gaps persist in our understanding of the full spectrum of AA's molecular mechanisms and in the development of effective therapeutic approaches. This review aims to comprehensively explore the immunological, genetic, epigenetic, and environmental factors underlying AA, with a focus on immune-mediated mechanisms. We also evaluate diagnostic approaches and recent advancements in assessing disease severity. Furthermore, the review discusses evolving therapeutic options, including traditional therapies, biologics, small-molecule agents, and emerging treatments. The academic value of this work lies in its synthesis of current knowledge on the multifaceted nature of AA, providing insights for future research and clinical practice. By elucidating the interconnected factors underlying AA, this review seeks to advance both understanding and management of this prevalent, clinically challenging disorder.

  • LETTER
    Ruiyang Ding , Xiaoke Ren , Qinglin Sun , Shiqian liu , Linyuan Huang , Zhiwei Sun , Junchao Duan
  • REVIEW
    Luying Yang , Fan Shi , Feng Cao , Le Wang , Jianzhen She , Boling He , Xiaoying Xu , Liang Kong , Bolei Cai

    Tissue repair represents a highly intricate and ordered dynamic process, critically reliant on the orchestration of immune cells. Among these, neutrophils, the most abundant leukocytes in the body, emerge as the initial immune responders at injury sites. Traditionally recognized for their antimicrobial functions in innate immunity, neutrophils now garner attention for their indispensable roles in tissue repair. This review delves into their novel functions during the early stages of tissue injury. We elucidate the mechanisms underlying neutrophil recruitment and activation following tissue damage and explore their contributions to vascular network formation. Furthermore, we investigate the pivotal role of neutrophils during the initial phase of repair across different tissue types. Of particular interest is the investigation into how the fate of neutrophils influences overall tissue healing outcomes. By shedding light on these emerging aspects of neutrophil function in tissue repair, this review aims to pave the way for novel strategies and approaches in future organ defect repair, regeneration studies, and advancements in tissue engineering. The insights provided here have the potential to significantly impact the field of tissue repair and regeneration.

  • HIGHLIGHT
    Hongquan Liu , Tianqi Wang , Jitao Wu
  • LETTER
    Sidi Yang , Kun Li , Qifan Zhou , Xumu Zhang , Deyin Guo
  • HIGHLIGHT
    Qi Weng , Qi Wu , Quan Zheng
  • HIGHLIGHT
    Peng Su , Mao Li , Fangfang Zhou
  • LETTER
    Jiang He , Tangmin Lai , Yuzu Zhao , Haonan Yang , Zheng Lei , Liu Zhou , Nan Li , Yu He , Wei Zhou , YongZhong Wu
  • ORIGINAL ARTICLE
    Rui Qiao , Yuanchen Liu , Qiyu Mao , Jiayan Li , Yinying Lu , Jialu Shi , Chen Li , Jizhen Yu , Jiami Gong , Xun Wang , Yuchen Shao , Lei Sun , Wenhong Zhang , Hongjie Yu , Hin Chu , Pengfei Wang , Xiaoyu Zhao

    The ongoing emergence of new variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the urgent need for developing antivirals targeting both SARS-CoV-2 variants and related sarbecoviruses. To this end, we designed novel trispecific antibodies, Tri-1 and Tri-2, engineered by fusing the single-chain variable fragments (scFvs) of a potent antibody (PW5-570) to the Fc region of “Knob-into-Hole” bispecific antibodies (bsAbs) composed of two distinct broad antibodies (PW5-5 and PW5-535). Compared with the parental antibodies, Tri-1 and Tri-2 displayed enhanced binding affinities to the receptor-binding domains of SARS-CoV, SARS-CoV-2 wild type, and Omicron XBB.1.16, with each arm contributed to the overall enhancement. Furthermore, pseudovirus neutralization assays revealed that Tri-1 and Tri-2 effectively neutralized all tested SARS-CoV, SARS-CoV-2 variants, and related sarbecoviruses (Pangolin-GD, RaTG13, WIV1, and SHC014), demonstrating potency and breadth superior to any single parental antibody. Consistently, Tri-1 and Tri-2 were found to effectively neutralize authentic SARS-CoV and SARS-CoV-2 variants with IC50 values comparable to or better than those of parental antibodies. Taken together, this study highlights the potential effectiveness of Tri-1 and Tri-2 as novel formats for harnessing cross-neutralizing antibodies in the development of multivalent agents to combat both current and future SARS-like coronaviruses.

  • REVIEW
    Van T. Hoang , Quyen Thi Nguyen , Trang Thi Kieu Phan , Trang H. Pham , Nhung Thi Hong Dinh , Le Phuong Hoang Anh , Lan Thi Mai Dao , Van Dat Bui , Hong-Nhung Dao , Duc Son Le , Anh Thi Lan Ngo , Quang-Duong Le , Liem Nguyen Thanh

    From the pioneering days of cell therapy to the achievement of bioprinting organs, tissue engineering, and regenerative medicine have seen tremendous technological advancements, offering solutions for restoring damaged tissues and organs. However, only a few products and technologies have received United States Food and Drug Administration approval. This review highlights significant progress in cell therapy, extracellular vesicle-based therapy, and tissue engineering. Hematopoietic stem cell transplantation is a powerful tool for treating many diseases, especially hematological malignancies. Mesenchymal stem cells have been extensively studied. The discovery of induced pluripotent stem cells has revolutionized disease modeling and regenerative applications, paving the way for personalized medicine. Gene therapy represents an innovative approach to the treatment of genetic disorders. Additionally, extracellular vesicle-based therapies have emerged as rising stars, offering promising solutions in diagnostics, cell-free therapeutics, drug delivery, and targeted therapy. Advances in tissue engineering enable complex tissue constructs, further transforming the field. Despite these advancements, many technical, ethical, and regulatory challenges remain. This review addresses the current bottlenecks, emphasizing novel technologies and interdisciplinary research to overcome these hurdles. Standardizing practices and conducting clinical trials will balance innovation and regulation, improving patient outcomes and quality of life.

  • REVIEW
    Jiayi Jin , Lijun Yue , Maoru Du , Feng Geng , Xue Gao , Yuming Zhou , Qianqian Lu , Xiaohong Pan

    Molecular hydrogen (H2), recognized as the smallest gas molecule, is capable of permeating cellular membranes and diffusing throughout the body. Due to its high bioavailability, H2 is considered a therapeutic gas for the treatment of various diseases. The therapeutic efficacy of hydrogen is contingent upon factors such as the administration method, duration of contact with diseased tissue, and concentration at targeted sites. H2 can be administered exogenously and is also produced endogenously within the intestinal tract. A comprehensive understanding of its delivery mechanisms and modes of action is crucial for advancing hydrogen medicine. This review highlights H₂’s mechanisms of action, summarizes its administration methods, and explores advancements in treating intestinal diseases (e.g., inflammatory bowel disease, intestinal ischemia–reperfusion, colorectal cancer). Additionally, its applications in managing other diseases are discussed. Finally, the challenges associated with its clinical application and potential solutions are explored. We propose that current delivery challenges faced by H2 can be effectively addressed through the use of nanoplatforms; furthermore, interactions between hydrogen and gut microbiota may provide insights into its mechanisms for treating intestinal diseases. Future research should explore the synergistic effects of H2 in conjunction with conventional therapies and develop personalized treatment plans to achieve precision medicine.

  • REVIEW
    Boqiang Tao , Xiaolan Li , Ming Hao , Tian Tian , Yuyang Li , Xiang Li , Chun Yang , Qirong Li , Qiang Feng , Hengzong Zhou , Yicheng Zhao , Dongxu Wang , Weiwei Liu

    Organoid technology, as an emerging field within biotechnology, has demonstrated transformative potential in advancing precision medicine. This review systematically outlines the translational trajectory of organoids from bench to bedside, emphasizing their construction methodologies, key regulatory factors, and multifaceted applications in personalized healthcare. By recapitulating physiological architectures and disease phenotypes through three-dimensional culture systems, organoids leverage natural and synthetic scaffolds, stem cell sources, and spatiotemporal cytokine regulation to model tissue-specific microenvironments. Diverse organoid types—including skin, intestinal, lung, and tumor organoids—have facilitated breakthroughs in modeling tissue development, drug efficacy and toxicity screening, disease pathogenesis studies, and patient-tailored diagnostics. For instance, patient-derived tumor organoids preserve tumor heterogeneity and genomic profiles, serving as predictive platforms for individualized chemotherapy responses. In precision medicine, organoid-guided multiomics analyses identify actionable biomarkers and resistance mechanisms, while clustered regularly interspaced short palindromic repeats-based functional screens optimize therapeutic targeting. Despite preclinical successes, challenges persist in standardization, vascularization, and ethical considerations. Future integration of artificial intelligence, microfluidics, and spatial transcriptomics will enhance organoid scalability, reproducibility, and clinical relevance. By bridging molecular insights with patient-specific therapies, organoids are poised to revolutionize precision medicine, offering dynamic platforms for drug development, regenerative strategies, and individualized treatment paradigms.

  • ORIGINAL ARTICLE
    Yonika A. Larasati , Gonzalo P. Solis , Alexey Koval , Marie-Céline François-Heude , Julie Piarroux , Agathe Roubertie , Ruihan Yang , Ying Zhang , Dezhi Cao , Christian M. Korff , Vladimir L. Katanaev

    GNAO1-associated disorders have a large spectrum of neurological symptoms, from early-onset developmental and epileptic encephalopathies (DEE) to late-onset movement disorders. First reported in 2013 and now identified in around 400 cases worldwide, this disease is caused by dominant, mostly de novo missense mutations in GNAO1, the gene encoding the major neuronal G protein Gαo. Being the immediate transducer of a number of neuronal G protein-coupled receptors, Gαo plays crucial functions in brain development and physiology. Here, we discover a novel mutation site in GNAO1, Cys225 mutated to Tyr or Arg in pediatric individuals from France and China (p.(Cys225Tyr) and p.(Cys225Arg), respectively), leading to severe early-onset DEE. Molecular investigations characterize the novel pathogenic variants as deficient in the interactions with guanine nucleotides and physiological cellular partners of Gαo, with reduced stability and plasma membrane localization and a strong neomorphic interaction with the chaperone Ric8A. Salts of zinc, emerging as a promising targeted therapy for GNAO1-associated disorders, impose a previously unseen effect on the mutant Gαo, accelerating the loss of its ability to interact with guanine nucleotides. Our study, combining clinical, cellular, molecular, and modeling approaches, describes deep insights into molecular etiology and treatment perspectives of the novel form of GNAO1-associated disorders.

  • LETTER
    Cheng Yang , Wei-Hua Chen , Jie Qian , Rong-Chong Huang , Jian-Jun Li
  • PERSPECTIVE
    Avnesh S. Thakor

    Precision Medicine is thought of as having two main pillars: Precision Diagnosis and Precision Therapy. However, for Precision Medicine to reach its full potential, a third pillar is needed that we propose to call Precision Delivery. In the laboratory, many therapies show great efficacy when tested directly with target cells. However, upon clinical translation, they are often given via intravenous or oral administration, resulting in their systemic distribution. To ensure therapies reach target sites at the correct therapeutic levels, they are often given at higher concentrations. However, this can be associated with off-target effects, side-effects, and unwanted interactions. Delivery strategies can help mitigate this by “spatially re-coupling” therapies in vivo with target cells. This review explains the concept of Precision Delivery, which can be thought of as three interconnected, but independent, modules: targeted delivery, microenvironment modulation, and cellular interactions. While locoregional approaches directly deliver therapies into target tissues through endovascular, endoluminal, percutaneous, and implantation techniques, microenvironment modulation technologies facilitate the movement of therapies across biological barriers and through tissue matrices, so optimized therapies can reach and interact with target cells. We highlight new innovations driving advances in Precision Delivery, while also discussing the considerations and challenges that Precision Delivery faces as it becomes increasingly integrated into treatment workflows.

  • LETTER
    Xue Yang , Dingming Li , Yihong Yang , Guicheng Zhao , Ying Shen
  • ORIGINAL ARTICLE
    Dongxue Hu , Qiaoyun Yang , Hongxu Xian , Minghao Wang , Hong Zheng , Karthik Babu Mallilankaraman , Victor C. Yu , Yih-Cherng Liou

    Mitochondrial homeostasis is essential for cell survival and function, necessitating quality control mechanisms to ensure a healthy mitochondrial network. Death-associated protein 3 (DAP3) serves as a subunit of the mitochondrial ribosome, playing a pivotal role in the translation of mitochondrial-encoded proteins. Apart from its involvement in protein synthesis, DAP3 has been implicated in the process of cell death and mitochondrial dynamics. In this study, we demonstrate that DAP3 mediates cell death via intrinsic apoptosis by triggering excessive mitochondrial fragmentation, loss of mitochondrial membrane potential (ΔΨm), ATP decline, and oxidative stress. Notably, DAP3 induces mitochondrial fragmentation through the Mitochondrial Rho GTPase 1 (Miro1), independently of the canonical fusion/fission machinery. Mechanistically, DAP3 promotes mitochondrial calcium accumulation through the MCU complex, leading to decreased cytosolic Ca2+ levels. This reduction in cytosolic Ca2+ is sensed by Miro1, which subsequently drives mitochondrial fragmentation. Depletion of Miro1 or MCU alleviates mitochondrial fragmentation, oxidative stress, and cell death. Collectively, our findings reveal a novel function of the mitoribosomal protein DAP3 in regulating calcium signalling and maintaining mitochondrial homeostasis.

  • HIGHLIGHT
    Junyi Wang , Xiang He , Guoping Li