+Advanced Search

  • Neurodegenerative diseases (NDs) are a wide variety of disorders characterized by the progressive and irreversible loss of neuronal structure and functions leading to cognitive impairments. The common types of NDs include Alzheimer’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and Parkinson’s disease. The sharing pathological hallmarks of these diseases are the aberrant aggregation and amyloid deposition. However, the underlying molecular mechanisms of protein misfolding and aberrant aggregation remain elusive. Amyloid protein is prone to aggregate from its native disordered monomeric state into well-ordered amyloid fibril state via nucleation-dependent polymerization mechanism, in which follows sigmoidal growth kinetics with three steps: lag phase, growth phase, and plateau phase. The formation and subsequent distribution of these pathological amyloid fibrils are closely related to the onset and progression of NDs. Additionally, the aberrant aggregation of these disease-associated proteins proceeds via liquid-liquid phase separation (LLPS) and liquid-to-solid phase transition (LSPT) leading to amyloid fibril formation in the condensed phase. The phase transition from liquid-like droplets or dynamic condensates to solid-like hydrogel or amyloids is intimately linked to the pathogenesis of several NDs. In this review, we discuss two typical pathways of amyloid fibrils formation. One route involves aggregation in the bulk solution environment, proceeding via nucleation and elongation steps to form amyloid fibrils. In this scenario, protein aggregation initiates with the nucleation step to form oligomeric nuclei. Then the nuclei serve as templates for the subsequent elongation step ultimately leading to the formation of amyloid fibrils. When sufficient fibrils have formed during self-assembly, the secondary nucleation is triggered to generate new species of oligomers and fibrillar aggregates. The other route of fibril formation occurs in the condensed phase through LLPS and LSPT to form amyloid aggregates and deposits. The occurrence of a phase separation leads to the liquid-like droplets formation during the early stage of aggregation. Over time, these dynamic biomolecular condensates gradually solidify and ultimately evolve into a hydrogel state enriched by amyloid aggregates through a phase transition process. Evidence indicates that pathological phase transitions are early events in the pathogenesis of several NDs. It should be noted that these two routes are not independent or mutually exclusive. They are interconnected and function cooperatively during aberrant aggregation. The pathological progression of NDs is closely related to the dominant aggregation pathway involved in aberrant aggregation. Moreover, the molecular mechanisms underlying the formation of pathogenic amyloid deposits are intricately linked to the structural and functional characteristics of aggregates. These aggregates may not only directly participate in fibrillization, but also indirectly promote the development of NDs by affecting the normal physiological cellular functions. Therefore, in-depth research on the structural and functional properties of both intermediates and fibrils is of great significance for understanding the molecular mechanisms of protein misfolding and aberrant aggregation. Overall, this paper reviews the amyloid deposition and pathological phase transitions in NDs. By delving into the molecular mechanisms of amyloid fibrillization, the aim is to better understand the pathogenesis of NDs, and to provide valuable insights into the development of therapeutic strategies targeting amyloid aggregation and aberrant phase transition.
    Citation
    LIANG Yu-Han, CHENG Wan-Ru, YANG Shuo, FENG Shuang, NIU Zheng.Nucleation-dependent Polymerization and Liquid-to-solid Phase Transition in Protein Aggregation[J].,2025,52(11):2689-2699.Export: BibTex EndNote
  • TAN Shu-Yi, ZHANG Jian
    2025, 52(11): 2025,52(11):2700-2716
    DOI: 10.3724/j.pibb.2025.0317
    In recent years, immunotherapy has become an excellent option for cancer patients, but most patients still face problems such as low response or drug resistance. Therefore, researchers conducted extensive studies on the reasons for the poor efficacy of immunotherapy. Eventually, it was found that the regulatory effect of abnormal expression of oncogenes and tumor suppressor genes on the tumor immune microenvironment is one of the important factors leading to the failure of immunotherapy to achieve the expected efficacy. It is well known that cancer is a kind of disease caused by the interaction between environmental and genetic factors, and the occurrence of cancer is mainly related to genetic alteration. Physiologically, the balance between oncogenes and tumor suppressor genes is crucial for DNA replication and proliferation regulation. However, under certain conditions, such as viral infection, chemical carcinogens or radiation, these genes may be mutated and eventually induce cancer. In addition, the combination of different gene mutations can also lead to significant differences among patients. For example, certain gene mutations are associated with the metastasis of cancer cells, while some are associated with the resistance of cancer cells to the attack of immune cells. Therefore, exploring the effects of different genetic alterations on the tumor microenvironment can help us better solve the problems in the process of clinical treatment and provide a theoretical basis for designing gene-targeted and personalized therapies. This review mainly summarizes the effects of common oncogenes and tumor suppressor gene mutations on immunosuppressive cells, anti-tumor immune effector cells and tumor-associated fibroblasts in the tumor microenvironment. Firstly, when the oncogene KRAS, c-Myc and EGFR are abnormally activated, cancer cell will secrete various cytokines and chemokines, thereby recruiting various immunosuppressive cells to the TME and causing exhaustion of CD8+ T and NK cells. It can also reprogram CAFs and eventually promote the development of cancer. Furthermore, similar phenomena occur after the inactivation of tumor suppressor genes. For example, cancer cells with inactivated PTEN genes will secrete large amounts of IL-33 and LOX to recruit macrophages and induce TAMs. Cancer cells can secrete a variety of microRNAs into the tumor microenvironment after p53 dysregulation. These mircoRNAs can reprogram CAFs and lead to epithelial-mesenchymal transition. Finally, we summarize the reversing effects of therapeutic interventions targeting mutant oncogenes or tumor suppressor genes (such as KRAS inhibitors, overexpression of p53 by mRNA, PI3Kβ inhibitors) on the immunosuppressive tumor microenvironment. Some of the results of their synergistic effects in combination with immunotherapy are also listed. Compared with monotherapy, the combination of either KRAS inhibitor or p53 mRNA nanomedicine with αPD-1 therapy resulted in more durable and potent anti-tumor effects. In summary, this review elucidates the regulatory and remodeling effects of genetic alterations in tumor cells on the tumor immune microenvironment, and analyzes the great potential of gene alteration intervention combined with immunotherapy. We hope it can provide theoretical basis and development strategy for precise cancer immunotherapy.
    Citation
    TAN Shu-Yi, ZHANG Jian.Regulatory Effects of Oncogenes and Tumor Suppressor Genes on Tumor Immune Microenvironment[J].,2025,52(11):2700-2716.Export: BibTex EndNote
  • Endometriosis (EM) and adenomyosis (AM) are chronic, estrogen-dependent gynecological disorders that significantly impair the quality of life and reproductive health of millions of women worldwide. Clinically, both conditions are characterized by dysmenorrhea, abnormal uterine bleeding, infertility, and high recurrence rates. Despite decades of research, their pathogenesis remains incompletely understood, and current therapeutic options are limited in both efficacy and long-term safety. Emerging studies have identified glycolytic metabolic reprogramming (GMR)—a shift from mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis—as a unifying and critical feature in the development and progression of EM and AM. In ectopic lesions, enhanced glycolysis supports cellular proliferation, survival, and adaptation to hypoxic microenvironments. Key glycolytic enzymes, including hexokinase 2 (HK2), phosphofructokinase-1 (PFK1), pyruvate dehydrogenase kinase (PDK), and lactate dehydrogenase A (LDHA), are markedly upregulated, whereas oxidative metabolism is suppressed, reflecting a Warburg-like metabolic phenotype. Notably, single-cell and spatial transcriptomic analyses reveal significant heterogeneity between EM and AM lesions. EM lesions often contain cell clusters co-expressing glycolytic and OXPHOS-related genes, suggesting metabolic flexibility. In contrast, AM tissues exhibit a more uniform, glycolysis-dominant profile, with preferential HK2 expression over HK1—potentially linked to defective repair of the endometrial basal layer. Multiple regulatory layers contribute to this glycolytic shift. Hypoxia-inducible factors (HIFs) act as upstream transcriptional activators in response to oxygen deprivation. Kinase cascades, such as those involving PIM2 and AURKA, enhance glycolytic enzyme activity via phosphorylation. Epigenetic mechanisms—including N6-methyladenosine (m6A) RNA modification and histone H3K18 lactylation—further stabilize glycolytic gene expression and reinforce metabolic reprogramming. These alterations form an integrated regulatory network that sustains high glycolytic flux in ectopic cells. Importantly, GMR profoundly affects the immune microenvironment. Lactate produced by glycolytic stromal cells promotes M2 macrophage polarization and impairs the function of cytotoxic T cells and dendritic cells, leading to immune evasion and chronic inflammation. Meanwhile, immune cells themselves undergo metabolic reprogramming, exhibiting increased dependence on glycolysis and diminished oxidative capacity. This bidirectional metabolic-immune feedback loop facilitates lesion persistence and disease progression. GMR is also closely linked to infertility in EM and AM. In the ovarian microenvironment, glycolytic imbalance leads to lactate accumulation in follicular fluid, negatively affecting oocyte quality and embryo development. In the endometrium, excessive glycolysis disrupts decidualization, angiogenesis, and immune tolerance—processes essential for implantation and pregnancy. Targeting glycolysis offers promising therapeutic potential. Small-molecule inhibitors such as dichloroacetate and meclozine target PDK and HK2, respectively. Natural compounds like cinnamic acid and protoberberine derivatives exhibit both anti-glycolytic and anti-inflammatory effects. Traditional Chinese medicine formulations, including Guizhi Fuling Wan, have shown efficacy in modulating metabolism, vascular remodeling, and fibrosis. Combination therapies, such as atorvastatin with resveratrol, may provide synergistic benefits by inhibiting both glucose uptake and lactate export. In conclusion, glycolytic metabolic reprogramming is a central mechanism linking inflammation, immune dysfunction, lesion progression, and reproductive failure in endometriotic diseases. Future research should focus on identifying metabolic subtypes, developing combined metabolic-immune therapies, and evaluating the safety of these treatments in reproductive-age women. These insights may pave the way toward personalized, mechanism-driven interventions for EM and AM.
    Citation
    DU Lin, WANG Mei-Ling, ZHOU Shuang-Shuang, FU Xian-Yun, SHI Wen-Jie, TAO Yi-Dan, ZHOU Hao-Xin.Glycolytic Hyperactivity in Endometriotic Diseases: From Molecular Mechanisms to Precise Interventions[J].,2025,52(11):2717-2728.Export: BibTex EndNote
  • LI Ru-Ru, ZHANG Ye, WEI Tao-Tao, ZHU Li
    2025, 52(11): 2025,52(11):2729-2748
    DOI: 10.3724/j.pibb.2025.0363
    Mitochondria are the most crucial energy-generating organelles in eukaryotic cells and serve as signaling hubs that orchestrate metabolism, redox balance, cell-fate decision and multiple forms of cell death. Mitochondria possess their own DNA (mtDNA), which is independent of the nuclear genome, yet encodes only 13 polypeptides, 22 tRNAs, and 2 rRNAs. The remaining >1 150 mitochondrial proteins are encoded by nuclear genes (nDNA), and the two genomes cooperate to preserve cellular homeostasis and proper function. Mitochondrial proteins are localized to the outer mitochondrial membrane (OMM), intermembrane space (IMS), inner mitochondrial membrane (IMM) or matrix, participating in oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, fission-fusion dynamics, and other processes indispensable for mitochondrial integrity. Mitochondrial quality control (MQC) is exerted largely by mitochondrial proteases, which selectively modulate protein activity and degrade misfolded or superfluous proteins. Among them, a group of mitochondrial ATPases associated with diverse cellular activities (AAA+ proteases) couple ATP binding and hydrolysis to protein unfolding and proteolysis, thereby regulating fusion protein maturation, respiratory-chain assembly, and mtDNA replication/transcription. Mutations or aberrant expression of these mitochondrial AAA+ proteases cripple mitochondrial architecture and function, precipitating a spectrum of severe neurological disorders. This review summarizes current knowledge on three paradigmatic mitochondrial AAA+ proteases, LONP1, YME1L1, and AFG3L2. We highlight their conserved Walker A/B motifs in the ATPase domain and hexameric architecture, yet emphasize divergent sub-mitochondrial topologies: LONP1 is soluble in the matrix, whereas YME1L1 and AFG3L2 are embedded in the IMM with catalytic domains facing IMS and matrix, respectively. These positional differences translate into distinct substrates and proteolytic strategies, enabling a division of labor and mutual complementation that cooperatively safeguards mitochondrial proteostasis. Pathogenic mutations linked to neurological disorders are mapped predominantly to the ATPase and the hydrolase/peptidase domains. Substitutions of the amino acid within these core domains can directly abolish ATP hydrolysis, substrate engagement or peptide cleavage, thereby crippling local MQC networks. Additional variants may disturb transcriptional, translational or post-translational regulation, altering protease stoichiometry and impairing compartmental balance. The subsequent cascade, mtDNA instability, respiratory-chain dysfunction, and aberrant mitochondrial dynamics, propagates stress signals that culminate in neuronal dysfunction and/or neurodegeneration. The mutational and clinical heterogeneity observed across cell types, developmental stages, and genetic backgrounds underscores the context-dependent fine-tuning of these AAA+ proteases. Deciphering how disease-associated variants rewire domain structure, catalytic cycle, and network-level crosstalk will therefore illuminate pathophysiologic mechanisms and guide precision therapeutic strategies.
    Citation
    LI Ru-Ru, ZHANG Ye, WEI Tao-Tao, ZHU Li.Structure and Function of Mitochondrial AAA+ Proteases and Their Roles in Neurological Disorders[J].,2025,52(11):2729-2748.Export: BibTex EndNote
  • Irisin, a myokine discovered in recent years, has been widely confirmed to exert cardioprotective effects. This review comprehensively elaborates on the molecular mechanisms of irisin in diabetic cardiomyocytes and its close associations with pathophysiological processes such as disordered glycolipid metabolism, oxidative stress, and autophagy. In terms of regulating glycolipid metabolism, irisin significantly improves energy metabolism in cardiomyocytes by activating the AMPK signaling pathway, thereby reversing diabetes-induced metabolic abnormalities. It promotes the browning of white adipose tissue (WAT), a process in which subcutaneous fat demonstrates a greater propensity to brown compared to visceral fat, thereby enhancing energy expenditure and exerting anti-inflammatory effects. These browned adipocytes secrete bioactive substances such as FGF and adiponectin, which further contribute to metabolic balance. Meanwhile, irisin reduces the glucolipotoxic burden on pancreatic β-cells: by modulating signaling pathways including PI3K/AKT and AMPK, it not only inhibits β-cell apoptosis but also improves their function and morphology. It enhances insulin secretion by regulating key proteins including Glut2, Glk, and Pdx1 through the AMPK pathway. Additionally, irisin accelerates the oxidation of free fatty acids (FFA) via activation of pathways such as PPARα, ameliorates insulin resistance, and thus optimizes the metabolic environment of cardiomyocytes. In the context of cellular stress regulation, irisin exhibits potent antioxidant properties. It not only directly counteracts the accumulation of reactive oxygen species (ROS) to alleviate oxidative damage but also inhibits ferroptosis by upregulating the MITOL/MARCH5 signaling axis, thereby helping to maintain mitochondrial homeostasis. Regarding endoplasmic reticulum stress (ERS), irisin downregulates key proteins including GRP78 and PERK, thus mitigating ERS-induced cardiomyocyte apoptosis and fibrosis—a protective mechanism that has also been validated in other diseases such as pancreatitis and osteoporosis. In maintaining the balance between autophagy and cell death, irisin sustains cellular homeostasis by coordinating both mitochondrial-targeted autophagy and non-selective autophagy. It promotes FUNDC1-mediated mitophagy to support mitochondrial turnover and ensure proper organelle function. At the same time, it suppresses excessive autophagy-induced cell damage through pathways such as PI3K/AKT/mTOR. In terms of apoptosis regulation, irisin downregulates pro-inflammatory factors (e.g., TNF-α, IL-6) and apoptosis-related proteins such as Caspase-3, while upregulating the anti-apoptotic protein Bcl-2. It inhibits cardiomyocyte apoptosis through multiple signaling pathways, including AMPK/mTOR and miR-19b/PTEN. In summary, irisin plays a crucial protective role in improving metabolic disorders, reducing cellular stress damage, and regulating cell death in diabetic cardiomyopathy (DCM) through multi-target and multi-pathway synergistic mechanisms. Its diverse actions provide an important theoretical basis and potential therapeutic targets for the clinical prevention and treatment of DCM. However, further research is needed to clarify its systemic effects, the safety of clinical interventions, and optimal treatment strategies to fully realize its therapeutic potential.
    Citation
    YAN Xue-Ru, ZHANG Yue-Jun, LI Jia-Yue, ZHANG Hao-Da, HE En-Peng.The Cellular Mechanism of Irisin in improving Diabetic Cardiomyopathy[J].,2025,52(11):2749-2758.Export: BibTex EndNote
  • Neurodegenerative diseases (NDs) are a group of disorders characterized by the progressive loss of neuronal structure and function, leading to clinical manifestations such as cognitive decline, motor dysfunction, and neuropsychiatric abnormalities. NDs encompass a range of conditions, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), etc. With the intensifying trends of global population growth and aging, the incidence of NDs continues to rise, yet no curative treatments are currently available. The blood-brain barrier (BBB) plays a crucial role in maintaining central nervous system (CNS) homeostasis by blocking harmful substances in the bloodstream from entering brain tissue. More than 98% of small-molecule drugs and nearly 100% of large-molecule therapeutics fail to cross the BBB and reach brain parenchyma. Ultrasound-targeted microbubble destruction (UTMD) is an emerging interdisciplinary technology integrating materials science and bioengineering, which combines the advantages of microbubble carriers with the physical properties of ultrasound. This innovative approach enables transient and reversible opening of the BBB, and enhancing drug delivery efficiency. Microbubbles (MB) are the core component of the UTMD system, consisting of two fundamental structural elements: a gaseous core and a biocompatible outer shell. The drug-loading capacity of MB has been significantly expanded, evolving from traditional chemotherapeutic agents to encompass nucleic acid drugs, macromolecular antibodies, and even traditional Chinese medicines. Concurrently, their drug-loading strategies have advanced from initial passive physical adsorption to active targeted delivery. UTMD possesses the following 4 biological advantages. (1) UTMD can transiently and reversibly enhance the permeability of cell membranes and blood vessels. The biocompatible shells commonly used in microbubbles can be metabolized by the body, posing no risk of long-term accumulation. (2) UTMD not only significantly improves drug delivery efficiency but also simultaneously serves as an ultrasound contrast agent and therapeutic carrier, achieving the integration of diagnosis and treatment. (3) UTMD technology offers dual advantages of spatial targeting and molecular targeting, allowing for precise drug delivery. (4) UTMD only requires conventional ultrasound equipment, and the raw materials for microbubble preparation are readily available with simple synthesis processes. Whether applied in diagnostics or treatment, the cost remains relatively low. The mechanism by which UTMD opens the BBB is primarily associated with cavitation effect and sonoporation effect. The cavitation effect induces mechanical stretching of both cellular membranes and capillary walls, creating transient, reversible channels that facilitate macromolecular drug passage, to enhance BBB permeability. Meanwhile, the sonoporation effect promotes drug penetration through dual mechanisms: (1) augmenting passive diffusion across biological barriers; (2) potentiating active transport processes. This synergistic action significantly elevates both local drug concentrations and therapeutic efficacy at target sites. The permeability of BBB is predominantly influenced by both microbubble characteristics and ultrasound parameters. Microbubble characteristics and ultrasound parameters are key factors affecting BBB permeability. By adjusting the composition of microbubbles and optimizing ultrasound parameters, effective BBB opening can be achieved while minimizing tissue damage, to regulate the dosage of drugs delivered to the brain parenchyma. Both preclinical investigations and clinical trials have consistently shown that UTMD holds significant therapeutic promise for NDs. This article outlines the fundamental properties of microbubbles and elucidates the potential mechanisms underlying UTMD mediated BBB opening. Furthermore, it systematically reviews recent advances in UTMD technology for the treatment of treating various NDs, aiming to provide a theoretical foundation and future directions for developing novel therapeutic strategies and drugs for NDs.
    Citation
    LI Ling-Yan, ZHENG Ruo-Quan, HU Huo-Jun, YOU Cheng-Cheng, YANG Yi, SHENG De-Qiao, ZHOU Jun, HUANG Yi-Ling.Ultrasound-targeted Microbubbles Destruction: a New Approach to The Treatment of Neurodegenerative Diseases[J].,2025,52(11):2759-2771.Export: BibTex EndNote
  • Schizophrenia is a severe psychiatric disorder characterized by positive symptoms (e.g., hallucinations), negative symptoms (e.g., social withdrawal), and cognitive impairments. Among these, cognitive impairment is a core feature that severely compromises patients’ social functioning and long-term prognosis. Antipsychotics, the first-line treatment for schizophrenia, are generally effective in managing positive symptoms. However, their efficacy in alleviating negative symptoms and cognitive deficits remains limited. Moreover, long-term use may lead to metabolic syndrome and extrapyramidal side effects. Consequently, non-pharmacological interventions have garnered increasing attention as alternative or adjunctive strategies for cognitive remediation in schizophrenia. In recent years, techniques grounded in neuroplasticity theory have advanced rapidly. These interventions aim to alleviate cognitive impairments by modulating neural circuits (e.g., enhancing prefrontal-hippocampal connectivity) and synaptic plasticity (e.g., modulating the BDNF/TrkB pathway) from multiple dimensions. Such approaches not only enhance cognitive function but also reduce medication-related adverse effects and improve treatment compliance. This article comprehensively reviews the clinical evidence and recent technological advances in non-pharmacological interventions targeting cognitive impairments in schizophrenia. The interventions discussed include cognitive remediation therapy (CRT), repetitive transcranial magnetic stimulation (rTMS), transcranial direct current stimulation (tDCS), electro-acupuncture (EA), aerobic exercise (AE), and light therapy (LT). CRT, the most extensively studied and evidence-based intervention, uses structured cognitive training tasks to enhance neuroplasticity and has consistently demonstrated efficacy in improving executive function and social cognition. Both rTMS and tDCS are non-invasive brain stimulation techniques that modulate cortical excitability and neural network connectivity. While rTMS has shown promise in improving working memory and attention—particularly in patients with prominent negative symptoms—its clinical efficacy remains inconsistent, likely due to variability in stimulation parameters and patient heterogeneity. In contrast, tDCS has demonstrated encouraging effects on working memory and attention with a relatively rapid onset, although optimal stimulation protocols have yet to be standardized. EA, which combines traditional acupuncture with electrical stimulation, has been shown to improve memory function, possibly through upregulation of brain-derived neurotrophic factor (BDNF) and enhanced cerebral blood flow. It may be especially useful in treatment-resistant cases. AE is a low-cost and widely accessible intervention that promotes hippocampal neuroplasticity and BDNF expression, thereby improving memory and attention. It is recommended as a foundational adjunctive therapy, particularly for patients with chronic schizophrenia. LT, although still experimental, has yielded promising results in animal models by modulating neuroinflammation and enhancing neurogenesis via the BDNF/CREB signaling pathway. However, clinical evidence remains limited, necessitating further large-scale trials to validate its efficacy and safety. In addition to reviewing individual interventions, this article highlights the potential of combination strategies—such as CRT combined with AE or rTMS—to produce synergistic cognitive benefits. Future directions include the development of personalized treatment protocols, early intervention during neurodevelopmental windows (e.g., adolescence), and the integration of biomarkers and neuroimaging to guide therapeutic decisions. This synthesis aims to provide clinicians and researchers with a comprehensive framework for advancing non-pharmacological cognitive rehabilitation in schizophrenia.
    Citation
    FENG Jia-Xin, XIE Yan-Hong, LI Yi, LIN Fo-Xiang, HUANG Min-Fang, WANG Qin-Wen, WANG Zheng-Chun.Non-pharmacological Treatments for Core Cognitive Impairment in Schizophrenia[J].,2025,52(11):2772-2787.Export: BibTex EndNote
  • Parkinson’s disease (PD), the second most common neurodegenerative disorder worldwide, presents significant heterogeneity in clinical manifestations, genetic background, and response to interventions. While conventional exercise therapies demonstrate benefits in alleviating motor and non-motor symptoms through mechanisms such as modulating α-synuclein aggregation, enhancing mitophagy, and reducing neuroinflammation, their efficacy varies considerably among individuals. This variability may stem from endogenous factors such as genetic background, clinical phenotypes, stages of pathological progression, as well as exogenous factors like the type, intensity, and frequency of movement. Thus, this review first discusses the necessity of precise exercise interventions for PD patients, focusing on the epidemiological burden, heterogeneity in disease mechanisms, and differences in intervention response (Why). Next, we systematically explain how to develop precise exercise intervention strategies by stratifying interventions based on genetic background, clinical phenotype, and disease stage, combined with technological aids (How). Genetically, mutations in genes such as GBA1, PRKN, PINK1, and SNCA dictate distinct molecular pathologies—including lysosomal dysfunction, impaired mitophagy, and α-synuclein aggregation—which necessitate tailored exercise regimens. For instance, patients with PRKN/PINK1 mutations may benefit from moderate-intensity endurance training to support mitochondrial biogenesis without exacerbating oxidative stress, whereas carriers of GBA1 mutations might require exercises focusing on enhancing lysosomal function and managing oxidative damage. Clinically, patients are stratified into tremor-dominant (TD) and postural instability/gait difficulty (PIGD) subtypes, which demand divergent exercise priorities: coordinative, rhythm-based activities like dance or Tai Chi for TD-PD to engage cerebellar circuits, versus targeted balance and strength training, potentially aided by virtual reality, for PIGD-PD to mitigate axial symptoms and fall risk. Furthermore, intervention strategies must evolve with disease progression: high-intensity exercise is prioritized in early stages to leverage neuroplasticity and potential disease modification, while mid- and late-stage management focuses on functional maintenance, fall prevention, and compensatory strategies, respectively. Critical to implementing this framework is the adoption of digital biomarkers via wearable technology (e.g., inertial sensors, smartwatches), which enables continuous, objective monitoring of gait, tremor, and physiological responses. This facilitates a closed-loop feedback system, allowing for the remote adjustment of exercise parameters (intensity, frequency, duration) in real-time, thus optimizing efficacy and ensuring safety. Finally, we detail how to configure exercise parameters through personalized adaptation (What), including exercise type, intensity, frequency and dose. Higher volumes of physical activity are associated with reduced PD risk and slower progression, though optimal thresholds remain incompletely defined. Aerobic exercise improves cardiovascular fitness and may aid clearance of pathogenic proteins; resistance training counters sarcopenia and bradykinesia; balance training reduces falls; and mind-body exercises (e.g., Tai Chi) integrate motor and cognitive components. Multimodal regimens are often most beneficial. High-intensity aerobic exercise appears particularly effective in early PD, enhancing neural connectivity and mitigating disease progression in randomized trials. Most evidence supports supervised sessions occurring 3-5 times per week, lasting 30-60 min, adapted to individual tolerance and disease stage. In conclusion, this narrative review outlines a comprehensive precision medicine framework for exercise intervention in PD, moving beyond symptomatic management towards targeting underlying pathophysiology. By stratifying patients based on genetic, phenotypic, and staging characteristics, and by leveraging digital technology for dynamic personalization, exercise therapy can be transformed into a more potent, individualized, and disease-modifying strategy. Future research must validate these biomarker-driven approaches in large-scale trials and establish definitive guidelines for translating precision exercise into clinical practice.
    Citation
    ZHOU Zi-Gui, YAN Min, WEN Xiao, WANG Hui, LIU Guo-Qiang, TIAN Xue-Wen.Multidimensional System of Precision Exercise Interventions for Parkinson’s Disease: Dynamic Regulation Based on Genetic Typing, Motor Subtypes, Clinical Staging, and Wearable Digital Biomarkers[J].,2025,52(11):2788-2801.Export: BibTex EndNote
  • Regulatory T cells (Treg cells) are a specialized subset of CD4+ T cells defined by expression of the lineage-specifying transcription factor FOXP3 and a potent capacity to maintain peripheral immune tolerance. The modern concept of Tregs was catalyzed by Shimon Sakaguchi’s identification of CD4+CD25+ suppressive T cells and subsequent work establishing FOXP3 as a central determinant of Treg cell development and function; together with landmark FOXP3 genetic discoveries by Mary E. Brunkow and Fred Ramsdell, these advances transformed understanding of immune homeostasis and were recognized by the 2025 Nobel Prize in Physiology or Medicine. Under normal physiological conditions, FOXP3+ Treg cells restrain autoreactive lymphocytes, prevent excessive inflammation, and shape antigen-presenting cell activity through contact-dependent pathways and suppressive cytokines, thereby protecting tissues from immune-mediated damage. Disruption of Treg abundance, stability, or suppressive capacity can therefore lead to immune dysregulation and disease. Over the past two decades, Treg cells have become a major focus of immunology because their roles are highly context dependent. In autoimmune and chronic inflammatory diseases, impaired Treg cell function or insufficient Treg activity contributes to loss of tolerance and persistent tissue injury, supporting therapeutic approaches designed to enhance Treg cell number, stability, and suppressive potency. In contrast, many cancers exploit Treg cells by promoting their expansion, activation, and recruitment into the tumor microenvironment (TME), where they blunt antitumor immunity by suppressing cytotoxic T-cell priming and effector function, limiting dendritic cell activation, and fostering immune escape. In both settings, immune checkpoint pathways critically influence Treg cell biology. Beyond PD-1/PD-L1 and CTLA-4, emerging checkpoints and costimulatory receptors, including TIGIT, TIM-3, LAG-3, and OX40, modulate Treg cell generation, stability, and suppressive programs, thereby shaping the balance between tolerance and immunity. Meanwhile, immunometabolic adaptations further tune Treg cell fitness and function in inflamed tissues and tumors; lipid utilization and mitochondrial programs, among other metabolic axes, enable Treg cells to persist in nutrient- and oxygen-restricted microenvironments, while microenvironmental stress can drive functional remodeling or fragility in a subset-dependent manner. In this review, we summarize the discovery and defining biological features of Tregs, highlight core suppressive mechanisms and regulatory circuits, and synthesize evidence for the dual roles of Treg cells in preventing autoimmunity yet enabling tumor immune evasion. We further outline current and emerging therapeutic strategies aimed at augmenting Treg cell activity to restore tolerance in autoimmune disease, or selectively depleting, functionally inhibiting, and reprogramming tumor-resident Treg cells to enhance cancer immunotherapy, including immune checkpoint blockade and combination approaches. Finally, we discuss how deeper insight into Treg heterogeneity, checkpoint control, and immunometabolic regulation may enable more precise Treg cell-directed interventions and inform next-generation immunotherapeutic combinations across immune-mediated and malignant diseases.
    Citation
    RIAZ Farooq, LIANG Ming-Wei, LI Yi-Kui, JIANG An-Mei, ZHANG Zhen-Zhen, ZHOU Zhi-Yi, FAN Zu-Sen, PAN Fan.Discovery of Regulatory T Cells and Their Prospective Therapeutic Applications[J]..Export: BibTex EndNote
  • Objective This study aimed to construct a cell co-culture microfluidic chip based on droplet microfluidic to investigate the influence of multicellular interactions in complex microenvironments on the sensitivity of anti-tumor drugs.Methods We constructed a droplet microfluidic chip consisting of 12 co-culture units, with each unit containing 4 microwells for holding cell droplets, enabling the co-culture of 4 types of cells. To evaluate whether the co-culture of multiple cell types can be achieved in the droplet microfluidic chip, as well as to observe and analyze the interactions between different cell types, we investigated the interaction between microenvironmental cells and tumor cells through cell co-culture experiments. To construct a stable co-culture system capable of evaluating drug sensitivity, we conducted diffusion experiments with blue ink and model drugs to investigate the ability of drugs to diffuse in the chip and be taken up by cells. To investigate the effect of the complex microenvironment on cellular drug sensitivity, we carried out cell co-culture experiments combined with drug treatment to explore the changes in the drug sensitivity of tumor cells in the presence of microenvironmental cells. To explore the reasons for drug resistance in tumor cells under co-culture conditions, we detected DNA double-strand break marker using immunofluorescence.Results Experiments on cell culture within droplets showed that cells in each droplet exhibited good proliferation ability and consistent cell status, laying a foundation for the co-culture of multiple kind of cells. Cell co-culture experiments showed that compared with the mono-culture group, the numbers of LoVo cells, HUVECs, and macrophages in the co-culture group increased significantly. This confirms that there are obvious interactions between cancer-associated fibroblasts (CAFs), endothelial cells (HUVECs), macrophages, and tumor cells in the microenvironment, which promotes cell proliferation in the co-culture chip. Experiments on blue ink diffusion showed that drugs could diffuse uniformly and effectively into wells in different directions within the chip. Experiments on the diffusion of doxorubicin (a model drug) demonstrated that identical cells in different wells exhibited consistent drug uptake capacity for the drug. Additionally, cell co-culture experiments combined with oxiliplatin treatment revealed that with the concentration of 80 μmol/L, the survival rate of LoVo cells cultured alone was only 25%, whereas it reached 96% under co-culture conditions. with the concentration of 160 μmol/L, the survival rate of LoVo cells cultured alone was merely 2%, while that under co-culture conditions was 50%. These results indicate that the complex microenvironment composed of CAFs, HUVECs, and macrophages significantly reduces the drug sensitivity of LoVo cells to oxaliplatin through intercellular interactions. The immunofluorescence results showed that the expression level of γH2AX in LoVo cells decreased under co-culture conditions.Conclusion Our study achieved co-culture of the main constituent cells of the tumor microenvironment and analysis of their drug sensitivity in a droplet microfluidic chip for the first time. The research found that crosstalk between different microenvironmental cells strongly affects the drug sensitivity of tumor cells to oxaliplatin, suggesting that targeting the interactions between tumor microenvironmental cells is an effective strategy to improve the efficacy of tumor therapy. Our study provides new methods and approaches for the efficacy evaluation of anti-tumor drugs and the screening of new drugs. In addition, the open structural design of the co-culture chip can be combined with various omics technologies to analyze the molecular characteristics of cells under co-culture and drug treatment conditions. This is expected to provide new methods and experimental evidence for elucidating the mechanisms of drug action and identifying novel drug targets in the context of the microenvironment.
    Citation
    ZHANG Xue-Tong, CHEN Shuo, FANG Jin.Establishment of a Multicellular Co-culture System Based on Droplet Microfluidic Chip for Analysis of Antitumor Drug Sensitivity[J]..Export: BibTex EndNote
  • Metal-organic frameworks (MOFs), a class of porous crystalline materials formed by the self - assembly of metal ions/clusters and organic ligands, exhibit broad application potential in the biomedical field due to their high specific surface area, precisely tunable pore structure, designable framework composition, and good biocompatibility. This paper traces the origin and development of MOFs, summarizes the contributions of the main promoters, and then systematically reviews the conventional synthesis and characterization methods of MOFs. It focuses on in - depth discussions around three application directions in the biomedical field: Firstly, in integrated cancer diagnosis and treatment, MOFs participate in tumor treatment through single or combined strategies of multiple treatment modalities such as chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, chemodynamic therapy, starvation therapy, and immunotherapy. Secondly, as carriers, MOFs achieve precise drug delivery and biomacromolecule loading by constructing pH - responsive carriers, GSH - responsive carriers, light - responsive carriers, and biomacromolecule carriers. Thirdly, in in vitro diagnosis, MOFs develop various biomarker detection methods to realize enzyme - linked immunosorbent assay and construct electrochemical and photochemical biosensors. Meanwhile, this paper deeply analyzes the key challenges that MOFs still face in clinical translation, including large - scale preparation, long - term stability, and biological safety assessment, and prospects the future development directions and application prospects.
    Citation
    GENG Fu-Kang, FANG Xiao-Cui, LEI Sheng-Bin, WANG Chen.Applications of Metal-organic Frameworks in The Field Of Biomedicine[J]..Export: BibTex EndNote
  • Circular RNAs (circRNAs) represent a distinct group of RNA molecules produced through back-splicing of precursor mRNAs. Their covalently closed structure, which lacks both a 5′ cap and a poly(A) tail, renders them highly resistant to exonucleolytic degradation and contributes to their remarkable intracellular stability. Although circRNAs were historically viewed as noncoding transcripts, accumulating evidence indicates that certain circRNAs can undergo translation under appropriate molecular contexts. Two major modes of noncanonical translation have been described so far: initiation mediated by internal ribosome entry sites (IRESs) and translation triggered by N6-methyladenosine (m6A) modification. These findings have broadened the traditional definition of noncoding RNA biology and suggest that circRNAs may contribute previously unrecognized elements to the cellular proteome. Peptides generated from circRNAs have been increasingly implicated in cancer biology. Depending on their molecular functions, these peptides may enhance malignant phenotypes—such as uncontrolled proliferation, motility, invasion, epithelial-mesenchymal transition, metabolic alteration, or drug resistance—or, conversely, exhibit inhibitory effects on oncogenic pathways. Their dual and context-dependent functions highlight the complexity of circRNA-mediated regulation and suggest that these translation products participate in multiple layers of tumor initiation and progression. In this review, we synthesize current knowledge regarding the molecular mechanisms that enable circRNAs to be translated, with particular attention to IRES-driven initiation, m6A-dependent regulation, ribosome accessibility, and the structural determinants required for translation competence. We further summarize well-characterized circRNA-encoded peptides and discuss how they influence tumor-associated signaling networks. In addition, we examine the potential translational applications of these peptides, including their value as diagnostic indicators, prognostic markers, or therapeutic entry points. Their inherent sequence stability, relative expression specificity, and detectability in clinical specimens make circRNA-derived peptides promising candidates for future biomarker and therapeutic development. Overall, circRNA translation research is reshaping our understanding of RNA function and offers new perspectives for studying tumor biology. We propose that expanding investigations into circRNA-encoded peptides will not only improve the mechanistic resolution of cancer research but may also pave the way for innovative strategies in precision oncology, including RNA-based therapeutics and peptide-targeting interventions.
    Citation
    XIANG Qiong, YANG Li-Chan, LI Zan, LING Yun.Translational Mechanisms of Circular RNAs and The Roles of Their Encoded Peptides in Tumor Initiation and Regulation[J]..Export: BibTex EndNote
  • Diabetic Nephropathy (DN) is the leading cause of End-Stage Renal Disease (ESRD) globally, representing a major global health burden with limited disease-modifying therapies. Podocyte injury serves as the core pathological hallmark of DN, and conventional treatments targeting metabolic disorders or hemodynamic abnormalities fail to reverse the progressive decline of renal function. Accumulating evidence over the past decade has established that high glucose-induced podocyte pyroptosis—a pro-inflammatory form of programmed cell death—is a key driving force in DN progression. Its core molecular mechanism hinges on the activation of the TXNIP-NLRP3 inflammasome axis. Under sustained hyperglycemic conditions, excessive Reactive Oxygen Species (ROS) are generated via pathways including the polyol pathway, advanced glycation end product (AGE) accumulation, and mitochondrial dysfunction. Concurrently, methylglyoxal (a glucose metabolite) mediates post-translational modification of Thioredoxin-Interacting Protein (TXNIP). These events collectively trigger the dissociation of TXNIP from Thioredoxin (TRX), a redox-regulating protein. The free TXNIP then translocates to the mitochondria, where it binds to The NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) (NLRP3) and promotes inflammasome assembly. This assembly activates Cysteine-Aspartic Acid Protease 1 (caspase-1), which cleaves Gasdermin D (GSDMD) to generate its N-terminal fragment (GSDMD-NT). GSDMD-NT oligomerizes to form membrane pores, leading to podocyte swelling, rupture, and the release of pro-inflammatory cytokines Interleukin-1β (IL-1β) and Interleukin-18 (IL-18). These cytokines amplify local inflammatory responses, induce mesangial cell proliferation, and accelerate extracellular matrix deposition, ultimately exacerbating glomerulosclerosis. MCC950, a highly selective NLRP3 inhibitor, exerts its therapeutic effects through a multi-layered mechanism: it binds to the NACHT domain (NAIP, CIITA, HET-E and TP1 Domain) of NLRP3 with nanomolar affinity, forming hydrogen bonds with key residues (Lys-42 and Asp-166) within the ATP-hydrolysis pocket to block ATP hydrolysis, thereby locking NLRP3 in an inactive conformational state. Additionally, MCC950 interferes with the protein-protein interaction between TXNIP and NLRP3 and regulates mitochondrial homeostasis to reduce ROS production. Preclinical studies have demonstrated that MCC950 dose-dependently reduces proteinuria, restores the expression of podocyte-specific markers (Nephrin and Wilms Tumor 1 Protein, WT1), and alleviates podocyte foot process fusion and glomerulosclerosis in both Streptozotocin (STZ)-induced type 1 diabetic models (characterized by absolute insulin deficiency) and db/db type 2 diabetic models (driven by insulin resistance). However, discrepancies in therapeutic outcomes exist across different models—some studies report exacerbated renal inflammation and fibrosis in STZ-induced models—which may stem from differences in disease pathogenesis, intervention timing (early vs. mid-stage disease), and dosing duration. Despite its promising preclinical efficacy, MCC950 faces significant translational challenges, including low oral bioavailability, insufficient podocyte targeting, potential hepatotoxicity, and drug-drug interactions with statins (commonly prescribed to diabetic patients for cardiovascular risk management). Furthermore, off-target effects such as the inhibition of carbonic anhydrase 2 have been identified, raising concerns about its safety profile. Nevertheless, its unique mechanism of action—directly blocking podocyte pyroptosis by targeting the TXNIP-NLRP3 axis—endows it with substantial translational value. In the future, strategies to overcome these barriers are expected to advance its clinical application: targeted delivery via nanocarriers (e.g., PLGA-PEG nanoparticles or nephrin antibody-conjugated systems) to enhance renal accumulation and podocyte specificity; precise patient stratification based on biomarkers such as serum IL-18 and renal TXNIP/NLRP3 expression to identify "inflammatory-phenotype" DN patients most likely to benefit; and combination therapy with Sodium-Glucose Cotransporter 2 (SGLT2) inhibitors—whose metabolic benefits synergize with MCC950"s anti-inflammatory effects. These approaches hold great potential to break through clinical translation bottlenecks, offering a novel, precise anti-inflammatory treatment option for DN and addressing an unmet clinical need for therapies targeting the inflammatory underpinnings of the disease.
    Citation
    ZHENG Hong, MO Zhong-Cheng, LIU Hang, PAN Xi-Zhang, WEI Bing.MCC950 Targeted Inhibition of TXNIP-NLRP3 Axis-mediated Podocyte Pyroptosis in Diabetic Nephropathy[J]..Export: BibTex EndNote
  • Cancer remains a leading cause of global mortality, necessitating the development of advanced therapeutic strategies with enhanced efficacy and reduced systemic toxicity. Among promising bioactive agents, lactoferrin (LF)—a multifunctional iron-binding glycoprotein abundantly found in mammalian milk and exocrine secretions—has garnered significant interest for its potent and multifaceted anti-cancer properties. This review provides a comprehensive analysis of the current understanding of LF"s role in oncology, encompassing its structural biology, diverse mechanisms of action, and groundbreaking advancements in its application through nano-engineering. LF exerts anti-tumor effects through multiple pathways, including extracellular action, intracellular action, and immune regulation. It demonstrates a remarkable affinity for cancer cell membranes, binding to overexpressed anionic components such as glycosaminoglycans and sialic acids, as well as to specific receptors including the Low-density Lipoprotein Receptor-related Protein 1 (LRP-1). This selective binding facilitates targeted uptake. Upon internalization, LF orchestrates a direct assault by inducing cell-cycle arrest in phases such as G0/G1 or S phase through the modulation of key regulators including cyclins, CDKs, and p53. Furthermore, it promotes programmed cell death via apoptotic pathways, involving caspase activation and downregulation of anti-apoptotic proteins such as survivin. A more recently elucidated mechanism is the induction of ferroptosis, an iron-dependent form of cell death characterized by overwhelming lipid peroxidation. Beyond direct cytotoxicity, LF acts as a potent immunomodulator. It enhances Natural Killer (NK) cell activity, modulates T-lymphocyte populations, and crucially reprograms Tumor-Associated Macrophages (TAMs) from a pro-tumor M2 state to an anti-tumor M1 state, thereby reversing the immunosuppressive tumor microenvironment (TME). The translation of LF"s potential has been significantly accelerated by nanotechnology. The inherent biocompatibility and natural tumor-targeting capabilities of LF make it an ideal platform for sophisticated drug-delivery systems. This review details various fabrication strategies for LF-based nanoparticles (NPs), including self-assembly, sol-oil emulsion, and electrostatic complexation, among others. Research demonstrates that nano-formulations not only protect LF from degradation but also enhance its bioactivity and anti-cancer potency. More importantly, LF NPs serve as versatile carriers for a wide array of therapeutic agents, including conventional chemotherapeutics, natural compounds, and imaging agents. These engineered systems enable synergistic therapy and facilitate site-specific delivery. Notably, the ability of LF to bind to receptors on the blood-brain barrier (BBB) has been leveraged to develop nano-systems for glioblastoma treatment. Other innovative designs utilize LF to modulate the TME—for instance, by alleviating tumor hypoxia to sensitize cells to radiotherapy and chemotherapy. Despite compelling pre-clinical evidence, the clinical translation of LF and its nano-formulations remains nascent. While early-phase trials have established a favorable safety profile for recombinant human LF, larger Phase III studies have yielded mixed results, underscoring the complexity of its action in humans. Key challenges include enhancing drug targeting, optimizing loading efficiency, ensuring batch-to-batch reproducibility, and achieving deep tumor penetration. Future research must focus on the rational design of next-generation LF-NPs. This entails developing standardized manufacturing protocols, engineering "smart" stimuli-responsive systems for targeted drug release in the TME, and constructing multi-targeting platforms. A concerted interdisciplinary effort is paramount to bridge the gap between bench and bedside. In conclusion, LF, particularly in its nano-engineered forms, represents a highly promising and versatile agent in the oncological arsenal, holding immense potential for precise and effective cancer therapy.
    Citation
    YUE Wen-Tian, HE Shu-Rong, AN Qin, ZOU Yun-Xia, DONG Wen-Wen, MENG Qing-Yong, ZHANG Ya-Li.Applications of Lactoferrin and Its Nanoparticles in Cancer Therapy[J]..Export: BibTex EndNote
  • Atherosclerosis (AS), the primary pathological contributor to cardiovascular diseases (CVDs), has increasingly affected younger populations due to modern dietary habits and sedentary lifestyles. Current diagnostic modalities, including ultrasound, MRI, and CT, primarily identify advanced lesions and inadequately evaluate plaque vulnerability, thereby hindering early detection. Conventional treatments, which involve long-term medications associated with side effects such as hepatic injury and surgical interventions that carry risks of restenosis and hemorrhage, underscore the urgent need for non-invasive, cost-effective early diagnostic methods and targeted therapies. Gut microbiota metabolites are pivotal in AS pathogenesis, with trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFAs) serving as functionally opposing biomarkers. TMAO is produced when gut bacteria, specifically Firmicutes and Proteobacteria, metabolize dietary choline and carnitine into trimethylamine (TMA), which the liver subsequently converts to TMAO via flavin-containing monooxygenase 3 (FMO3); TMAO is then excreted in urine. Variability in TMAO levels is influenced by marine food consumption and FMO3 modulation, which can be affected by genetics, age, and diet. Mechanistically, TMAO exacerbates AS by disrupting cholesterol metabolism, inducing endothelial dysfunction through the elevation of reactive oxygen species (ROS) and pro-inflammatory cytokines such as IL-6, and reducing nitric oxide levels. Additionally, TMAO activates NF-κB and NLRP3 pathways while enhancing platelet reactivity. Clinically, elevated TMAO levels correlate with early AS and serve as predictors of mortality in patients with stable coronary artery disease (CAD) and acute coronary syndrome (ACS), as well as major adverse cardiovascular events (MACE) in stroke patients. Conversely, SCFAs—namely acetate, propionate, and butyrate—are produced by gut bacteria such as Akkermansia muciniphila and Faecalibacterium prausnitzii through the fermentation of dietary fiber. These metabolites exert anti-AS effects: acetate aids in maintaining metabolic homeostasis; propionate protects endothelial function and reduces plaque area; and butyrate fortifies intestinal barriers while suppressing inflammation. Furthermore, SCFAs cross-regulate bile acid metabolism, thereby influencing TMAO levels, and antagonize the pro-inflammatory and lipid-disrupting effects of TMAO. The use of TMAO and SCFAs as standalone biomarkers is constrained by limitations. TMAO lacks specificity, while SCFA levels fluctuate based on gut microbiota and dietary intake. Traditional AS risk assessment tools, which include clinical indicators, imaging techniques, and single biomarkers such as CRP, LDL-C, and ASCVD scores, overlook gut metabolism and demonstrate inadequate performance in younger populations. This review advocates for an "antagonistic-complementary" combined strategy: utilizing acetate and TMAO for early AS, propionate and TMAO for progressive AS, and butyrate and TMAO for advanced AS, addressing endothelial dysfunction, lipid deposition, and plaque stability/thrombosis risk, respectively. For clinical application, standardization of detection methods is crucial; liquid chromatography-mass spectrometry (LC-MS) is the gold standard, necessitating a unified sample pretreatment protocol, such as extraction with 1% formic acid in methanol. Additionally, dried blood spots (DBS) facilitate non-invasive testing, provided that dietary controls are implemented prior to detection, including a 12-hour fast and avoidance of high-choline and high-fiber foods. Existing challenges encompass the absence of standardized systems, limited large-scale validation, and ambiguous interactions with conditions such as hypertension. The authors" team has previously established connections between gut metabolites and AS, including the reduction of TMAO as a preventive measure for AS, thereby reinforcing this proposed strategy. Future research should prioritize standardization, the development of machine learning-optimized models, validation of interventions, and the exploration of multi-omics-based "gut microbiota-metabolite-vascular" networks. In conclusion, the combined detection of TMAO and SCFAs offers a novel framework for AS risk assessment, facilitating early diagnosis and targeted interventions while enhancing the integration of gut metabolism into cardiovascular disease management.
    Citation
    SHI Zhi-Chao, TIAN Xu-Ping, CHEN Si-Yi, LIU Shi-Guo.The Prospect of Trimethylamine N-oxide Combined With Short-chain Fatty Acids in Atherosclerosis Risk Prediction[J]..Export: BibTex EndNote
  • Deciphering how the brain enables humans to interact, coordinate, and learn from one another remains one of the most compelling challenges in contemporary cognitive neuroscience. Social interaction is a dynamic, reciprocal process. Over the past decade, hyperscanning research has consistently identified Inter-Brain Synchronization (IBS) as a neural signature accompanying successful cooperation, communication, joint attention, and social learning. However, the correlational nature of these findings leaves a critical question unresolved: Does IBS cause better social interaction, or does it merely reflect it? While traditional hyperscanning paradigms are powerful in revealing inter-brain neural dynamics "in the wild," they cannot on their own determine the direction of causality. This gap has motivated the emergence of Multibrain Stimulation (MBS)—a new generation of causal inference tools designed to actively manipulate neural coupling across individuals. MBS leverages non-invasive transcranial electrical stimulation (tES) to modulate neural activity simultaneously in two or more interacting brains. Unlike conventional tES applied to a single individual, MBS employs coordinated stimulation parameters, such as synchronized waveforms or matched frequencies, to directly perturb the neural mechanisms underlying social interaction. By providing an exogenous, precisely controlled intervention on IBS, MBS satisfies interventionist criteria for establishing causal relationships: researchers can test whether modifying inter-brain synchrony leads to predictable changes in behavior, communication, or shared understanding. This capability represents a fundamental methodological shift, transforming interpersonal neuroscience from a largely descriptive discipline into one capable of mechanistic inquiry. The biophysical underpinnings of MBS vary depending on the specific modality used. Transcranial alternating current stimulation (tACS) functions through cross-brain entrainment: when two individuals receive oscillatory currents matched in frequency and phase (e.g., theta-, beta-, or gamma-band stimulation), their endogenous neural rhythms tend to align with the exogenous signal and, consequently, with each other. This alignment effectively instantiates principles of the Communication Through Coherence (CTC) framework, which posits that coherent oscillations optimize information exchange by synchronizing periods of excitability across neural populations. Meanwhile, transcranial direct current stimulation (tDCS) exerts its influence by altering the excitability of targeted cortical regions in a polarity-dependent manner, thereby tuning the computational readiness of social-cognitive hubs such as the temporoparietal junction, superior temporal cortex, or inferior frontal gyrus. A growing body of empirical evidence demonstrates that such manipulations yield robust behavioral effects. In joint motor tasks, in-phase tACS enhances interpersonal coordination by aligning motor preparation dynamics, reducing temporal variability, and enabling individuals to anticipate each other"s actions more effectively. In communication and social learning contexts, MBS targeting high-order integrative regions promotes conceptual alignment, accelerates knowledge transfer, and supports more efficient encoding of shared representations. Notably, the effects of MBS often persist beyond the stimulation period, suggesting short-term plasticity in cross-brain networks. Post-stimulation improvements in synchronization and coordination indicate that MBS may temporarily recalibrate the neural architecture underlying social interaction. However, these benefits exhibit strong parameter specificity—precise phase relationships (e.g., 0° in-phase versus 180° anti-phase) and frequency matching are essential for generating reliable behavioral outcomes. Taken together, MBS represents a transformative step toward establishing the causal principles of human sociality and offers a new avenue for probing how multiple brains become functionally aligned during interaction.
    Citation
    CHEN Han-Lin, LI Qi, LI Yuan-Yuan, PAN Ya-Feng.Multibrain Stimulation and Its Modulation on Social Interaction: A Transcranial Electrical Stimulation Approach[J]..Export: BibTex EndNote
  • The 2025 Nobel Prize in Physiology or Medicine was awarded to Mary E. Brunkow, Fred Ramsdell and Shimon Sakaguchi in recognition of their groundbreaking contributions to unraveling the mechanisms of peripheral immune tolerance. Regulatory T cells (Tregs), as the core components maintaining peripheral immune tolerance, exhibit high plasticity and heterogeneity. Dysregulation of Treg function is closely associated with autoimmune diseases, tumor progression, and transplant rejection. Forkhead box protein P3 (FOXP3) is a key transcription factor that controls the development and function of Tregs. This review discusses the classification of Tregs into thymic-derived Tregs (tTregs), peripherally induced Tregs (pTregs), and in vitro-induced Tregs (iTregs). It also elaborates on how Treg cells exert their inhibitory functions through multiple pathways, including the secretion of inhibitory factors, metabolic interference via competitive uptake of IL-2, and direct cell-cell contact.induced Tregs (iTregs). It also elaborates on how Treg cells exert their inhibitory functions through multiple pathways, including the secretion of inhibitory factors, metabolic interference via competitive uptake of IL In recent years, significant advances have been made in Treg and FOXP3 research, progressively deepening our understanding of Treg plasticity. Investigations have revealed their capacity to adapt and acquire features of effector T helper cell subsets—such as Th1, Th2, and Th17—under specific microenvironmental cues. This plasticity also poses challenges for therapeutic interventions, as Tregs can potentially lose their suppressive function and acquire pro-inflammatory properties, thereby exacerbating disease pathology. Furthermore, the concept of tissue-specific Treg specialization has emerged, highlighting distinct functional subsets resident in organs such as the gut, adipose tissue, and tumors. For instance, gut-resident Tregs maintain tolerance to commensal bacteria and dietary antigens, while tumor-infiltrating Tregs promote immune evasion by suppressing anti-tumor immunity. Concurrently, studies on the metabolic and epigenetic regulation of Tregs, including post-translational modifications of FOXP3 such as acetylation and ubiquitination, have uncovered intricate layers of control over their stability and function. Building upon these fundamental insights, this review synthesizes FOXP3-targeted therapeutic strategies. These encompass approaches to enhance Treg function in autoimmune diseases and transplantation, including adoptive cell therapies and pharmacological interventions. Conversely, strategies to antagonize Treg-mediated immunosuppression in oncology, such as immune checkpoint blockade, are discussed. Notably, the development of programmable engineered Tregs represents a particularly promising frontier for achieving antigen-specific immune modulation with enhanced precision and efficacy. However, the field of Treg research continues to grapple with several complex challenges. The deeper, underlying regulatory networks governing Treg biology remain incompletely understood. A comprehensive resolution of Treg heterogeneity is still lacking, and significant hurdles exist in maintaining the stability and function of Tregs during in vitro expansion and culture. Furthermore, the precision and efficacy of translating these findings into clinical applications require substantial improvement. Consequently, both the development of Treg-targeting pharmacological agents and the refinement of Treg-based cellular therapies demand more profound exploration. The ultimate goal is to overcome these obstacles and achieve transformative, breakthrough clinical outcomes in the foreseeable future.
    Citation
    JJIANG Huang-Hao, FAN Jing-Yuan, PENG Cheng, LI Bin.Regulatory T cells and FOXP3: Milestones and Cutting-edge Breakthroughs in Peripheral Immune Tolerance[J]..Export: BibTex EndNote
  • This paper presents a comprehensive exploration of the IPDPS teaching concept—a framework built upon the 5 core principles of interdisciplinarity, practicality, diversity, process-oriented, and soul-forging—and its systematic implementation in the general education course "Biology in Daily Life" at Sun Yat-sen University. Developed over 8 years of iterative practice, this educational model is designed to address critical challenges in cultivating top innovative talents within higher education. It specifically targets the overcoming of disciplinary barriers, the disconnection between academia and industry, the limitations of one-way knowledge transmission, the rigidity of traditional evaluation systems, and the lack of value guidance. The curriculum is innovatively structured around four life-centric modules—birth, aging, illness, and food—which seamlessly integrate cutting-edge advancements in life sciences with interdisciplinary knowledge, making complex biological concepts accessible and relevant to students from diverse academic backgrounds. Pedagogically, the course employs a rich array of teaching methods to activate student engagement and foster higher-order thinking skills. These include case-based learning? driven by real-world problems, multi-sensory interactive experiences?, storytelling? to illustrate scientific discovery processes, and contrastive analysis? of ethical dilemmas in science. A cornerstone of the implementation is a multi-tiered practical teaching system, encompassing mandatory in-class experiments, optional social investigations, corporate visits, and face-to-face sessions with industry leaders. This structure ensures learning extends from the classroom to real-world societal and industrial contexts. A significant reform is the shift from a summative to a process-oriented evaluation system. This system diversifies assessment methods and incorporates multiple evaluators, including teacher assessment, self-assessment, and structured peer review using detailed rubrics. This approach aims to stimulate intrinsic motivation, foster a growth mindset, and provide a more holistic measurement of student development. Fundamentally, the course deeply integrates value-shaping elements? into its fabric. By incorporating themes of national identity, scientific spirit, bioethics, and cultural confidence through specific cases, the course forges students" sense of social responsibility and ethical reasoning, ensuring their innovative capacities are guided by a strong moral compass. Assessment data from 2017 to 2024 demonstrates significant positive outcomes. Course satisfaction ratings have shown a remarkable increase, rising from 80.5% to 96.8%. Survey data from 245 students (2021–2024) indicates that the course effectively broadens interdisciplinary horizons, enhances independent thinking and problem-solving abilities, and successfully integrates knowledge acquisition with capacity building and value orientation. The course has successfully functioned as an "initial incubator and screening mechanism"? for identifying and nurturing talented individuals, with some students even shifting their academic focus to biology as a result. In conclusion, the "Biology in Daily Life" course, underpinned by the IPDPS framework, provides a replicable and scalable paradigm for educational innovation in cultivating elite innovators. It represents a successful model for achieving the organic unity of knowledge impartation, ability cultivation, and value shaping in higher education. Future work will focus on optimizing differentiated content design for diverse student backgrounds, deepening practical teaching experiences, and establishing long-term tracking mechanisms for learning outcomes.
    Citation
    ZHU Ying, LI Lian, YANG Jin-E.Implementing The IPDPS Teaching Concept in “Biology in Daily Life” General Education Course for Cultivating Elite Innovators at Universities[J]..Export: BibTex EndNote
  • Regulatory T cells (Treg cells) have reshaped modern immunology by establishing the conceptual and mechanistic foundation of peripheral immune tolerance. Since the pioneering identification of CD4?CD25? suppressive T cells by Shimon Sakaguchi and the subsequent discovery of the lineage-defining transcription factor Forkhead box P3 (Foxp3) by Mary E. Brunkow and Fred Ramsdell, Treg cells have been recognized as indispensable guardians of immune homeostasis. These advances collectively clarified that central tolerance alone is insufficient to eliminate all self-reactive lymphocytes, and peripheral tolerance—critically mediated by Treg cells—serves as a second barrier preventing pathological autoimmunity. Contemporary research has therefore expanded the functional and therapeutic significance of Treg cells across the fields of autoimmunity, cancer, transplantation, and tissue repair. Treg cells originate from two major developmental pathways: thymus-derived Treg (tTreg) cells, which arise from high-affinity self-reactive TCR interactions in the thymus, and peripheral Treg (pTreg) cells, which are induced in mucosal and other peripheral tissues via antigen stimulation under tolerogenic cytokine cues such as IL-2 and TGF-β. Their differentiation is orchestrated by a multilayered transcriptional and epigenetic network within the Foxp3 locus, including CNS0–CNS3 elements that integrate TCR, cytokine and environmental signals to support lineage stability. Treg cells are identified by a combination of surface and intracellular markers—CD25, CD127low/?, CTLA-4, GITR, TNFR2, CD39/CD73, and Foxp3—although marker specificity varies with context, activation state, and species. Their notable heterogeneity enables Treg cells to adopt Th1-, Th2-, Th17- or Tfh-like programs through transcription factors such as T-bet, GATA3, RORγt and Bcl6, thereby permitting precise suppression of corresponding effector responses. Tissue-resident Treg subsets in adipose tissue, skin, skeletal muscle and the CNS have emerged as highly specialized regulators that integrate local metabolic and stromal signals, contributing not only to immunosuppression but also to tissue regeneration. Mechanistically, Treg cells maintain tolerance through three synergistic strategies: (1) secretion of suppressive cytokines (IL-10, TGF-β, IL-35) and cytotoxic mediators (granzyme B, perforin); (2) cell-contact–dependent interactions via CTLA-4, PD-1/PD-L1, and LAG-3 to limit dendritic cell maturation and T-cell activation; and (3) metabolic regulation including IL-2 consumption, adenosine production via CD39/CD73, cAMP transfer through gap junctions, and adaptation to hypoxic or nutrient-restricted microenvironments. Dysregulation of Treg cell quantity or function contributes directly to pathogenesis across a spectrum of diseases. In autoimmune diseases such as type 1 diabetes, systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis, impaired Foxp3 stability, epigenetic abnormalities, defective IL-2 signaling or inflammatory cytokine exposure undermine Treg suppressive capacity, facilitating excessive autoreactive T- and B-cell activation. In contrast, within the tumor microenvironment, Treg cells are often enriched through chemokine axes such as CCL22–CCR4 and reinforced by interaction with myeloid-derived suppressor cells and tumor-associated macrophages. Their enhanced metabolic fitness and suppressive phenotype enable tumors to evade immune destruction. In transplantation, Treg cells are essential for promoting graft tolerance, restraining effector T-cell activation, and facilitating tissue repair after injury. Rapid therapeutic progress has been driven by Treg-based immunomodulation. Polyclonal Treg adoptive transfer has demonstrated safety and preliminary efficacy in type 1 diabetes, autoimmune disorders, solid-organ transplantation, and graft-versus-host disease. Gene-engineered Treg therapies, including antigen-specific CAR-Treg and TCR-Treg platforms, offer superior precision and stability, enabling targeted suppression at disease sites. Additional strategies—including low-dose IL-2 therapy, small-molecule modulation, and selective depletion of intratumoral Treg using antibodies against CCR4, CCR8, CTLA-4 or CD25×TIGIT bispecifics—further expand the translational landscape. Collectively, advances in Treg biology—from lineage ontogeny and molecular regulation to specialized functions and therapeutic engineering—highlight Treg cells as central orchestrators of immune equilibrium. Continued integration of single-cell multi-omics, systems immunology and gene-editing technologies is expected to accelerate the development of highly specific, durable and safe Treg-centered therapies, ultimately enabling precision control of immune tolerance in autoimmunity, transplantation and cancer.
    Citation
    XIAO Teng, CHEN Meng-Yu, YI Lei, XIONG Wei, WANG Fu-Yan.Treg Cells and Peripheral Immune Tolerance: From Discovery to Precise Immune Regulation[J]..Export: BibTex EndNote
  • Objective Pioneer transcription factors (PTFs) possess the unique ability to recognize and bind their target DNA sequences within compacted nucleosomal DNA, thereby initiating chromatin opening and gene expression. They play pivotal roles in fundamental biological processes such as embryonic development, cellular reprogramming, and tumorigenesis. The specific regulatory mechanism by which nucleosomal rotational positioning governs PTF-nucleosome interactions remains inadequately elucidated. This study aims to systematically investigate the role of the rotational orientation of motifs in PTF-nucleosome binding.Methods We employed a DNA deformation energy model to predict the rotational positioning of DNA on nucleosomes. We analyzed high-throughput in vitro data from the NCAP-SELEX assay, which profiles the binding landscapes of numerous transcription factors to nucleosomal DNA. For in vivo analysis, we integrated genome-wide binding data (ChIP-seq) and nucleosome positioning data (MNase-seq) for eight well-characterized pioneer factors (OCT4, SOX2, KLF4, GATA4, MYOD1, FOXA1, CEBPA, and ASCL1) in human cells. Binding motifs were classified as "TF-bound" if they overlapped with ChIP-seq 峰s and "TF-unbound" otherwise. DNA bendability profiles and Fast Fourier Transform (FFT) analysis were used to assess rotational positioning patterns around these motif sites. This analytical framework was further applied to specific biological contexts, including cellular reprogramming from IMR90 fibroblasts to induced pluripotent stem cells (iPSCs) and the differentiation of human embryonic stem cells (hESCs) to human neuroectodermal cells (hNECs).Results Our in vitro analysis revealed a strong dependence of transcription factor binding on the rotational orientation of TF-binding motifs. For SOX7, the unbound motifs at specific enrichment 峰s exhibited a rotational phase clearly opposite to that of the SOX7-bound motifs. Similarly, analysis of P53 binding sequences confirmed that successful binding in vitro correlated with model-predicted exposure of the DNA minor groove at the motif center, consistent with P53"s binding mode. Genome-wide in vivo analysis of the eight PTFs showed that their DNA binding motifs were generally associated with DNA sequences exhibiting significant 10-bp periodicity in bendability, suggesting an inherent potential for nucleosome association. Crucially, for most factors (except ASCL1), the average rotational positioning preferences were remarkably similar between TF-bound and TF-unbound motifs. This indicates that, at a global genomic level, rotational positioning is not the primary determinant dictating whether a nucleosomal motif is bound by its cognate PTF in vivo. This phenomenon persisted during cellular reprogramming (IMR90 to iPSC), where the rotational positioning of OSKM factor motifs bound versus unbound in nucleosomal regions showed no significant overall difference. Interestingly, during hESC differentiation to hNECs, SOX2 binding sites underwent comprehensive reprogramming. In hNECs, the rotational positioning of nucleosomal SOX2-bound motifs was significantly different and, unexpectedly, opposite to the general preference observed in hESCs and for unbound motifs in hNECs, suggesting a cell context-dependent rewiring of binding mechanisms.Conclusion This study suggests a distinction in the role of DNA rotational positioning in TF-nucleosome binding between in vitro and in vivo environments. While rotational positioning critically governs the binding efficiency of factors like SOX7 and P53 in simplified in vitro systems, PTFs in vivo appear to overcome this steric hindrance at the binding interface. The ability of PTFs to bind nucleosomal motifs, even when key interaction surfaces are partially buried, might stem from their unique structural properties (e.g., intrinsically disordered regions, DNA distortion/binding domains), nucleosome breathing which transiently exposes DNA, and potential cooperativity with other factors. Our results highlight the unique capacity of pioneer factors to drive chromatin openness through mechanisms beyond rotational positioning.
    Citation
    LIU Guo-Qing, GUO Xing-Yue, CANG Jing, ZHANG Zhi, LIU Guo-Jun.Differential Role of Rotational Positioning in Pioneer Transcription Factor Binding to Nucleosomes In vivo vs. In vitro[J]..Export: BibTex EndNote
  • Methamphetamine (METH) addiction is a severe and increasingly prevalent neuropsychiatric disorder for which current diagnostic and therapeutic approaches remain limited and predominantly symptom-oriented. Exercise, as a safe, accessible and cost-effective non-pharmacological intervention, has emerged as a promising strategy to ameliorate METH-induced neurotoxicity and addiction-related behaviors. Growing evidence indicates that these benefits are closely linked to the regulation of exercise-induced biomarkers, defined as molecular indicators whose expression or activity is dynamically altered during or after physical activity. This review focuses on the core regulatory role of exercise-induced biomarkers in METH addiction and systematically summarizes their involvement in key neurobiological pathways, outlining molecular pathological mechanisms such as dysregulation of dopamine, glutamate and GABA neurotransmitter systems, neuroinflammation and oxidative stress, and epigenetic remodeling, and emphasizing how these processes converge on changes in candidate biomarkers in the brain and periphery. On this basis, the review describes how exercise modulates neural plasticity, neurotransmitter systems, inflammation and oxidative stress through biomarkers such as brain-derived neurotrophic factor (BDNF), exerkines, inflammatory cytokines, metabolites and non-coding RNAs, with particular attention to neurotrophic and immune-related markers, microRNAs and other epigenetic regulators that can reverse METH-induced synaptic and structural abnormalities and promote recovery of cognitive and emotional functions. Advances in high-throughput omics technologies, including transcriptomics, metabolomics and multi-omics integration, are summarized to illustrate the screening and identification of key exercise-responsive biomarkers. Studies in METH-addicted animal models have revealed differentially expressed genes, signaling pathways (e.g., PI3K-Akt, mTOR, Wnt) and core nodes such as NFKBIA and CXCL12 that may mediate the protective effects of exercise. The review further discusses the potential of exercise-mediated biomarkers as objective indicators for diagnosis, dynamic monitoring of therapeutic efficacy and patient stratification. Multi-gene diagnostic models based on peripheral samples (e.g., hair follicles, blood) demonstrate how biomarker panels can distinguish non-recovered, almost-recovered and healthy individuals, providing a molecular basis for staging METH use disorder and evaluating the impact of exercise interventions. The temporal dynamics of biomarker changes before and after exercise are highlighted, underscoring the value of longitudinal monitoring of factors such as BDNF, immune-related genes and circulating microRNAs to capture treatment-relevant windows of plasticity. In addition, the underlying molecular basis of exercise as an adjunct therapy and gene-targeted exercise strategies that leverage individual biomarker and gene expression profiles to optimize exercise prescriptions are summarized. Current conceptual and technical challenges are outlined, including heterogeneity of biomarker responses, individual variability, assay sensitivity and specificity, and gaps between preclinical findings and clinical application, together with future directions for integrating exercise with multi-omics, artificial intelligence-assisted biomarker discovery and, prospectively, gene-editing-based interventions. Particular emphasis is placed on the need to standardize exercise protocols, incorporate stage-specific and sex-sensitive designs, and combine exercise with pharmacotherapy and psychosocial rehabilitation in real-world clinical settings across diverse healthcare systems. Overall, this review aims to provide a comprehensive and integrated mechanistic framework and updated theoretical support for the application of exercise-mediated biomarkers in the diagnosis, therapeutic effect monitoring and personalized intervention of METH addiction, and to offer new and clinically relevant insights into the development of precision medicine strategies for substance use disorders.
    Citation
    HE Jin-Ke, ZHANG Xue-Jie, XU Ji-Sheng, LI Xue.Exercise-induced Biomarkers in Methamphetamine Addiction: Molecular Mechanisms and Clinical Implications[J]..Export: BibTex EndNote
  • Objective Osteoporosis is a progressive metabolic bone disorder characterized by reduced bone mass and microarchitectural deterioration, leading to increased skeletal fragility and susceptibility to fracture. Conventional diagnostic and risk-assessment approaches, such as dual-energy X-ray absorptiometry (DXA) and the FRAX? algorithm, remain limited because they rely primarily on bone mineral density (BMD) and a restricted set of clinical factors, failing to capture the multidimensional determinants of bone strength. This study aimed to develop and validate a deep learning-based multi-dimensional feature fusion model that integrates heterogeneous biological, structural, functional, and genetic information to improve the early identification of osteoporosis and enhance fracture risk prediction.Methods A total of 12 856 participants were aggregated from three major data repositories: the International Osteoporosis Foundation database, a clinical research database on osteoporosis, and a large-scale medical informatics dataset. A unified data-extraction protocol was applied to ensure cross-database harmonization, followed by quality control, variable standardization, and missing-data handling using multiple imputation by chained equations (MICE). A multimodal deep learning framework was constructed to integrate six categories of features: BMD measurements, quantitative bone microarchitecture parameters, bone turnover biomarkers, established clinical risk factors, osteoporosis-related genetic polymorphisms, and sensor-derived balance and gait metrics. A multi-task learning strategy was adopted to simultaneously predict osteoporosis status and 10-year fracture probability. Model training used five-fold cross-validation, and external validation was conducted in an independent clinical cohort. Model performance was benchmarked against DXA alone and the FRAX tool.Results In the internal test cohort, the proposed model achieved an AUC of 0.936 (95% CI: 0.927-0.945), with a sensitivity of 87.5% and a specificity of 91.2%, significantly outperforming DXA alone (AUC=0.889) and FRAX (AUC=0.842) (both P<0.05). External validation yielded an AUC of 0.918 (95% CI: 0.905-0.931) and demonstrated strong calibration (Brier score=0.087). SHAP analyses revealed that, beyond BMD, key predictors included trabecular separation, serum C-terminal telopeptide of type I collagen, balance-related metrics, gait speed, and specific SNPs within the RANKL and VDR loci. A simplified model incorporating only BMD, clinical features, and bone turnover markers preserved high accuracy (AUC=0.917), underscoring its feasibility for resource-limited clinical environments.Conclusion The deep learning-based multi-dimensional feature fusion model markedly enhances the precision and individualization of osteoporosis assessment compared with traditional tools. By integrating biological, structural, metabolic, genetic, and functional dimensions of bone health, the model provides a comprehensive representation of skeletal integrity and robustly improves both diagnostic accuracy and fracture risk prediction. Its strong generalizability across demographic subgroups highlights its clinical applicability. This work offers a promising direction for developing next-generation intelligent decision-support systems that may meaningfully improve osteoporosis screening, risk stratification, and preventive care.
    Citation
    WANG Chao-Ya, MENG Chao.Research on Multi-dimensional Feature Fusion Model for Osteoporosis Risk Assessment Based on Deep Learning[J]..Export: BibTex EndNote
  • Ischemic stroke (IS) accounts for approximately 80% of all stroke cases and is a leading cause of death and long-term disability worldwide. Its core pathological mechanism involves the interruption of cerebral blood flow, leading to neuronal cell death and ischemic tissue necrosis in the brain, which is associated with multiple molecular processes including apoptosis, inflammation, and oxidative stress. This review systematically discusses the classification of HDACs, the mechanisms of action of HDAC inhibitors, and their multiple effects in inhibiting cell apoptosis, regulating neuroinflammation, repairing the blood-brain barrier, and improving cognitive function following IS. HDACs function by removing acetyl groups from histone lysine residues, leading to chromatin condensation and gene silencing. The HDAC family is classified into four classes: class I (HDAC1, 2, 3, 8), class IIa (HDAC4, 5, 7, 9), class IIb (HDAC6, 10), and class IV (HDAC11), with class III being the NAD?-dependent sirtuins. Histone deacetylase inhibitors (HDACi) exert significant neuroprotective effects following ischemic stroke through a multi-target, multi-pathway synergistic mechanism. The core mechanisms include inhibition of neuronal apoptosis, regulation of neuroinflammation, protection of the blood-brain barrier (BBB), and improvement of cognitive impairments (PSCI). HDACi regulate gene expression epigenetically by upregulating genes such as p21/CIP1, leading to cell cycle arrest, while also modulating apoptosis-related proteins by inhibiting pro-apoptotic signaling pathways, thereby reducing neuronal cell death. In terms of neuroinflammation, HDACi suppress NF-κB and activate Nrf2 pathways, decreasing the release of pro-inflammatory cytokines and preventing the pro-inflammatory polarization of microglia and macrophages, thus modulating the inflammatory response. Regarding BBB protection, HDACi regulate the expression and restoration of tight junction proteins such as Occludin and Claudin-5, while inhibiting the release of destructive factors like MMP-9, alleviating vasogenic edema, and maintaining BBB integrity. Furthermore, HDACi promote the transcription of neurotrophic factors and synaptic-associated genes, enhancing neuroplasticity and repairing neuronal networks, ultimately improving cognitive functions. Therefore, HDACi demonstrate great potential as a multifaceted therapeutic strategy for ischemic stroke. HDAC inhibitors (HDACis) represent a powerful multi-target therapeutic approach that transcends the limitations of traditional thrombolytic therapies. HDACis represent a powerful multi-target therapeutic approach that transcends the limitations of traditional thrombolytic therapies, which are hampered by a narrow time window and risks of reperfusion injury. Histone acetylation is increased by HDACis, which relaxes chromatin and reactivates protective gene transcription. Their selectivity and chemical structure are used to classify them. Trichostatin A (TSA) and sodium butyrate (SB), a short-chain fatty acid, are examples of broad-spectrum inhibitors that are effective in lowering infarct volume and reducing neuroinflammation. More selective inhibitors, including tubastatin A (HDAC6-selective) and entinostat (class I-selective), may have fewer adverse effects while increasing efficacy. By suppressing apoptosis by modifying the p53, Bcl-2, and JNK pathways, reducing neuroinflammation by blocking NF-κB and NLRP3 activation, preserving the integrity of the blood-brain barrier by strengthening tight junction proteins, and promoting synaptic plasticity, neurogenesis, and the expression of neurotrophic factors like BDNF, these inhibitors provide neuroprotection through a variety of interrelated mechanisms.Despite their great potential, HDACis" clinical translation is fraught with difficulties, mostly because of non-selective inhibition-related adverse effects such as hepatotoxicity and gastrointestinal problems with valproic acid (VPA). In order to accomplish targeted delivery to the brain, future research is consequently shifting toward the development of highly selective inhibitors, refining dosing regimes, and utilizing cutting-edge drug delivery technologies like nanoparticles. In summary, the development of effective neuroprotective and neurorestorative treatments for IS may be greatly aided by a nuanced, spatiotemporally accurate understanding of HDAC activities and the judicious use of subtype-selective HDACis.
    Citation
    PAN Feng-Yuan, XU Yi-Wei, ZOU Xin-Yu, CHEN Jing.The Molecular Mechanisms of HDACi in Regulating Ischemic Stroke[J]..Export: BibTex EndNote
  • Parkinson’s disease (PD), the second most prevalent neurodegenerative disorder worldwide after Alzheimer’s disease, is pathologically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta and the abnormal intracellular aggregation of α-synuclein into Lewy bodies. Traditionally, the clinical symptoms of PD have focused on motor dysfunction, which includes characteristic signs such as resting tremor, rigidity, bradykinesia, and postural instability. However, increasing evidence from both clinical and basic research suggests that the clinical presentation of PD is highly diverse, with neuropsychiatric complications representing a significant and unavoidable aspect of the disease"s overall burden. From the perspective of clinical phenotypes, the range of neuropsychiatric symptoms associated with PD is extensive, primarily including depressive disorders, generalized anxiety, apathy, impulse control disorders, and cognitive impairments related to executive function and memory. Notably, emotional and cognitive dysfunctions often manifest years prior to the onset of motor symptoms. This clinical observation indicates that the pathological processes of PD may originate within the non-motor circuits of the central nervous system (CNS), particularly in neural networks closely linked to emotional regulation and cognitive function. As one of the human body"s most lipid-rich organs, the CNS comprises lipids that account for approximately 50%-60% of the dry weight of brain tissue. These lipid molecules serve not only as structural components but also actively participate in the formation of cell membrane phospholipid bilayers, myelin sheath insulation layers, and various signal transduction complexes. From a functional perspective, lipids not only provide the structural foundation necessary for maintaining neuronal membrane fluidity, synaptic plasticity, and ion channel activity, but also act as essential molecules in energy metabolism, signal transduction, and epigenetic regulation. Notably, the frontal cortex—particularly its evolutionarily specialized prefrontal cortex (PFC)—functions as the brain"s "executive center for cognition and emotion". This region is pivotal for higher cognitive functions, including working memory, decision-making, and behavioral inhibition, as well as for the complex regulation of emotions, such as reward and risk assessment. This region displays an exceptionally high synaptic density and is abundant in structural lipids, including unsaturated fatty acids and cholesterol, which makes it particularly vulnerable to disturbances in lipid metabolism. In PD research, lipid imbalance has become a central focus. As investigations progress, the importance of lipid metabolic pathways becomes increasingly apparent. Simultaneously, pharmacological therapies aimed at lipid regulation show considerable efficacy in addressing cognitive and emotional deficits associated with PD. In light of this, the present study utilizes bioinformatics analysis to identify differentially expressed genes in the peripheral blood of PD patients, demonstrating significant enrichment in processes such as chronic depression, cholesterol metabolism, fatty acid metabolism, AMPK signaling pathways, and insulin resistance. Expanding on this groundwork, the present review systematically explores the connections between dysregulated lipid metabolism and metabolic reprogramming in cognitive and emotional impairments associated with PD. Through the analysis findings, intervention approaches focusing on various fundamental pathological pathways such as neuroinflammation, mitochondrial dysfunction, imbalance in lactate homeostasis, and disrupted protein homeostasis are suggested. These proposals provide innovative perspectives for advancing mechanistic investigations and therapeutic advancements targeting cognitive and emotional disorders in PD.
    Citation
    LIU Xiao-Qian, Lü Meng-Lin, KOU Xian-Juan.The Mechanism of Lipid Metabolic Reprogramming in Cognitive and Emotional Impairments of Parkinson’s Disease[J]..Export: BibTex EndNote
  • Spinal cord injury (SCI) is a highly disabling trauma of the central nervous system, characterized by a complex pathological process involving intertwined multiple mechanisms. Key pathological events include excessive activation of neuroinflammation, oxidative stress injury, neuronal apoptosis, autophagic dysfunction, and energy metabolism imbalance, which severely disrupt the integrity of spinal cord neural function and significantly reduce patients" quality of life. Currently, clinical neurorepair strategies for SCI have limited efficacy and are difficult to achieve synergistic intervention targeting multiple pathological links. Therefore, exploring novel core therapeutic targets and precise intervention regimens has become an urgent need in this field. The Sirtuins family (SIRT1–SIRT7), as NAD?-dependent deacetylases, play a central role in critical biological processes such as cellular metabolism regulation, immune homeostasis maintenance, and stress injury repair, and have been identified as potential intervention targets for neurological diseases. This review systematically summarizes the cellular localization and core biological functions of each member of the Sirtuins family, with a focus on their regulatory roles and molecular mechanisms in the pathological process of SCI: SIRT1, 3, 5, and 6 inhibit the excessive activation of the NF-κB pathway and block NLRP3 inflammasome assembly through deacetylation modification, thereby participating in the regulation of neuroinflammation after SCI; meanwhile, they alleviate oxidative stress injury in spinal cord tissues by activating the Nrf2 antioxidant pathway and enhancing the activity of antioxidant enzymes such as SOD and NADPH, forming a "anti-inflammatory-antioxidant" synergistic protective effect. SIRT7 delays neuronal apoptosis by promoting DNA damage repair and inhibiting apoptotic signaling pathways. SIRT3 and SIRT5 target mitochondrial function, improve mitochondrial energy metabolism by regulating the modification status of enzymes involved in the tricarboxylic acid cycle and oxidative phosphorylation, and restore autophagic homeostasis by modulating the acetylation levels of FOXO3a and AMPK, providing metabolic support for neural repair. We summarize that a variety of natural Chinese herbal components (e.g., resveratrol, matrine) and synthetic compounds (e.g., SRT1720, AGK2) can influence the pathological progression of SCI by targeting and regulating members of the Sirtuins family. We propose that Sirtuins-targeted combination therapeutic strategies (e.g., combined with stem cell transplantation, neurotrophic factor supplementation, or antioxidant intervention) are expected to break through the limitations of single therapies and enhance the repair effect of SCI through multi-mechanism synergistic actions. In conclusion, the Sirtuins family exhibits critical mechanisms of action and potential intervention value in the pathophysiological process of SCI. This review summarizes and prospects novel Sirtuins-targeted therapeutic strategies, aiming to provide new insights for basic research and clinical translation in this field.
    Citation
    DU Hai-Lin, ZHANG Jian, LI Hong-Ru, CUI Yin-Jie, ZHENG Chen-Guang.Mechanisms and Therapeutic Prospects of The Sirtuins Family in Spinal Cord Injury[J]..Export: BibTex EndNote
  • As oncologic therapies continue to advance, the overall survival of cancer patients has markedly increased. Nevertheless, virtually every anticancer treatment modality is accompanied by some degree of cardiotoxicity. Epidemiological data indicate that approximately 30 % of cancer survivors ultimately die from cardiovascular disease. Among the cardiotoxic agents, the anthracycline doxorubicin (DOX) is the most widely used; it effectively suppresses a variety of malignant tumors—including breast cancer, lymphoma, and acute leukemia—but its cardiac toxicity limits further escalation of clinical dosing. Literature reports identify a cumulative dose of ≥250 mg/m2 as the threshold of high risk, with roughly 25 % of patients receiving DOX developing varying degrees of myocardial injury; severe cases progress to heart failure. Even at cumulative doses below the traditional safety limit, some patients exhibit cardiac dysfunction after the first administration, suggesting that cardiotoxicity is not solely a linear function of dose. DOX related cardiotoxicity can be classified as acute (hours to days after administration), sub acute (weeks to months), and chronic/late onset (years later). Most patients initially exhibit only mild reductions in left ventricular ejection fraction (LVEF) or subtle abnormalities in global longitudinal strain (GLS), often without symptoms. Recently, cardiac biomarkers (cTn, NT proBNP) combined with high sensitivity echocardiography (speckle tracking) have been recommended for monitoring high risk individuals, enabling detection of subclinical injury before overt LVEF decline. Currently, several preventive and therapeutic approaches are used in clinical practice, which can be summarized into the following four points: (1) dose limitation and administration strategies: fractionated low dose regimens, liposomal encapsulation, or continuous infusion lower peak plasma concentrations, thereby reducing cardiac exposure; (2) pharmacologic prophylaxis: β blockers (e.g., carvedilol) and ACE inhibitors/ARBs have shown protective effects on LVEF in some randomized trials, though results remain inconsistent and require larger confirmatory studies; (3) metabolic targeted interventions: animal experiments indicate that activation of PPARα or supplementation with L carnitine restores fatty acid oxidation and improves ATP generation, suggesting metabolic modulators as promising cardioprotective candidates; (4) lifestyle modifications: regular aerobic exercise up regulates mitochondrial biogenesis genes (PGC-1α) and reduces reactive oxygen species (ROS) production; small clinical studies have demonstrated a potential benefit in attenuating cTnT elevation. However, DOX-induced cardiotoxicity has not been effectively controlled, indicating that the core mechanism underlying DOX-related cardiac toxicity remains unidentified. Cardiomyocytes are high energy demand cells, and metabolic dysregulation is considered a central component of DOX induced cardiotoxicity. DOX disrupts myocardial metabolic balance through several interrelated pathways. (1) Oxidative stress and mitochondrial damage: DOX generates abundant ROS within cells, leading to mitochondrial membrane potential loss, lipid peroxidation, and iron accumulation, which suppress electron transport chain activity and markedly reduce ATP synthesis efficiency. (2) Autophagy dysregulation: DOX interferes with autophagic flux, preventing the clearance of damaged mitochondria and further aggravating apoptosis and inflammatory responses. (3) Inflammation and cytokine release: oxidative stress activates NF-κB, up-regulating pro inflammatory cytokines such as TNF-α and IL-6, creating a chronic inflammatory microenvironment that weakens myocardial contractility. (4) Epigenetic modifications: studies have shown that DOX alters DNA methylation and histone acetylation patterns in cardiomyocytes, affecting the expression of key metabolic genes (e.g., PGC-1α, CPT-1) and further inhibiting fatty acid β oxidation. These mechanisms collectively lead to suppressed fatty acid oxidation and compensatory up regulation of glycolysis, manifested by an elevated lactate/pyruvate ratio, accumulation of medium chain acyl carnitines, and a pronounced decline in ATP production. The resulting energy deficit precipitates left ventricular contractile dysfunction and, ultimately, heart failure. Despite extensive basic and clinical research on DOX cardiotoxicity, a unified risk assessment model and precise interventions targeting metabolic disturbances remain lacking. This review systematically summarizes recent progress on DOX induced cardiotoxicity and highlights that impairment of myocardial energy metabolism is a central mechanism of injury, thereby deepened our understanding of how impaired myocardial energy metabolism drives DOX induced injury, we can move toward safer chemotherapy protocols that achieve "cure cancer without harming the heart".
    Citation
    LIU Jia-Wei, JIA Tong-Xin, WU Jia-Zhen, SU Wen-Hua, GU Dan, DAN Ju-Hua.Does Doxorubicin Cause Heart Damage by Interfering With Heart Energy Metabolism?[J]..Export: BibTex EndNote
  • The inflammatory response is the foundation and a critical component of innate immunity. It serves as a vital defense mechanism, enabling the body to rapidly recognize and resist the invasion of foreign pathogenic microorganisms through a spontaneous immune reaction. Through pattern recognition receptors (PRRs), the host can effectively identify pathogen-associated molecular patterns (PAMPs) from microbes like bacteria and viruses, as well as damage-associated molecular patterns (DAMPs) released by injured cells. This allows for swift identification and resistance against pathogenic invasions, fulfilling a cellular surveillance function. As one of the most important protein complexes in innate immunity, the NLRP3 inflammasome—a large multi-protein complex—is among the most extensively studied inflammasomes. It assembles in response to pathogenic invasion or other danger signals and is crucial for the processing and release of pro-inflammatory mediators. This process helps the body distinguish between “self” and “non-self” and plays a significant role in both inflammatory and antiviral responses, thereby maintaining the host’s internal homeostasis. However, under certain conditions, immune regulation can become dysregulated, leading to an inflammatory response that is either too weak or too strong. This imbalance between pro-inflammatory and anti-inflammatory states can ultimately result in disease and tissue damage. Notably, not all viral infections activate the inflammasome. The activation mechanism of the NLRP3 inflammasome remains unclear and is even a subject of debate. On one hand, viruses are recognized by the host’s innate immune system, which can activate the NLRP3 inflammasome to mobilize immune and inflammatory responses for antiviral defense. Upon viral infection, the host receptor protein NLRP3 recognizes inflammatory signals, recruits the adapter protein ASC, and forms an inflammasome complex with pro-caspase-1. This triggers a cascade of activation events that initiate the innate immune response. Strategies involved in this process include altering intracellular and extracellular ion concentrations, affecting host cell energy metabolism, and directly interacting with components of the NLRP3 inflammasome to regulate its activation. On the other hand, viruses have evolved multiple strategies to inhibit NLRP3 inflammasome activation and evade immune responses. These include regulating NLRP3 ubiquitination and degradation, inhibiting the assembly and activation of the NLRP3 inflammasome, and modulating its effector functions. Furthermore, while NLRP3 inflammasome activation upon viral infection helps clear the virus and is crucial for antiviral defense, viruses can also evade this immune mechanism to facilitate their own replication and proliferation. A deeper understanding of the interplay between inflammasome activation and viral replication will contribute to the precise and effective prevention and treatment of currently incurable viral diseases. Therefore, this article will focus on the complex interactions between viral infection and the NLRP3 inflammasome. It will review recent advances in understanding virus-induced NLRP3 inflammasome activation and the immune evasion strategies viruses employ by modulating NLRP3 inflammasome activity, with the ultimate goal of fundamentally controlling viral replication in the host. In-depth research in this area will not only enhance our understanding of viral pathogenesis but also provide new strategies for clinical antiviral therapy and drug development.
    Citation
    BAI Guang-Ye, CHEN Deng-Jin, ZHANG Lei, ZHANG Qian, LIU Kai-Dong, HAO Hai-Yu, LI Peng, YI Fu-De, LI Jing-Lin, CHEN Shan, HAO Xiao-Jing.The Crosstalk Between Viral Infection and The NLRP3 Inflammasome[J]..Export: BibTex EndNote
  • The Alternative Lengthening of Telomeres (ALT) is a homology-directed repair (HDR)-based mechanism that maintains telomere length independently of telomerase by hijacking the canonical double-strand break (DSB) repair machinery. In ALT-positive cells, a RAD51-, MUS81-, and BLM-dependent recombination cascade copies telomeric tracts from sister chromatids, extrachromosomal telomeric circles (t-circles), or inter-chromosomal templates, thereby restoring a functional TTAGGG repeat array. This process is characterized by a distinct molecular signature:(1) chronic replication stress, manifested by elevated ATR–CHK1 signaling, R-loop accumulation, and fragile telomere phenotypes;(2) clustering of telomeric chromatin into ALT-associated PML bodies (APBs), which serve as SUMO-dependent recombination hubs enriched for SLX4–SLX1, MRE11–RAD50–NBS1, and FANCD2 complexes; and (3) global chromatin remodeling, marked by the eviction of histone H3.3 and its chaperones ATRX/DAXX, derepression of the long non-coding RNA TERRA, and acquisition of constitutive heterochromatin marks (H3K9me3/H4K20me3) along with the facultative heterochromatin mark H3K27me3. Together, these changes establish a chromatin environment permissive for homologous recombination. Importantly, these alterations are not merely passive by-products but are functionally required for homology search, strand invasion, and resolution of recombination intermediates. This is supported by CRISPR screens identifying ATRX, DAXX, and the SUMO E2 enzyme UBC9 as essential ALT fitness genes. While 85%–90% of human cancers re-express telomerase reverse transcriptase (TERT), the remaining 10%–15% are telomerase-null and rely exclusively on ALT for immortality. ALT tumors are enriched in osteosarcomas, glioblastomas, pancreatic neuroendocrine tumors, and aggressive soft-tissue sarcomas. In telomerase-negative somatic cells, progressive telomere shortening during each S phase eventually reaches a critical length, triggering a persistent DNA damage response (DDR) at chromosome ends. This activates the p53–p21 and p16INK4A–Rb tumor suppressor pathways, driving cells into stable replicative senescence. Although this telomere-length-dependent senescence acts as a potent barrier to malignant progression, recent single-cell analyses reveal that senescent fibroblasts and epithelial cells transiently display ALT-like features—such as accumulation of telomeric γH2AX/53BP1 foci, formation of APB-like PML condensates containing SUMOylated TRF1 and TRF2, and intermittent TERRA upregulation. These observations suggest that telomerase-negative tumors and senescent cells share a recombination-permissive chromatin state. Although senescent cells do not achieve net telomere elongation—likely due to intact p53/p16 checkpoints restraining unscheduled HDR—transient ALT activation may enable rare clonal escape. This further implies that ALT operates not only as a tumor-cell survival pathway but also as a protective mechanism against environmental stress. Indeed, spontaneous immortalization of TERT-/- fibroblasts in vitro is preceded by stochastic ALT induction, indicating that stochastic recombination at dysfunctional telomeres can overcome senescence barriers and initiate malignant transformation. Consistent with this model, whole-genome sequencing of ALT-positive tumors frequently identifies early driver mutations in TP53, ATRX, and DAXX, which disable replicative-senescence checkpoints while simultaneously enhancing telomeric HDR. Here, we synthesize the convergent molecular features of ALT tumors and senescent cells, highlighting:(1) replication stress as a common initiating cue, (2) SUMO-dependent phase separation as a platform for telomere-templated recombination, and (3) epigenetic erosion of ATRX/DAXX-mediated heterochromatin as a rate-limiting step. Finally, we discuss therapeutic implications: (1) pharmacological inhibition of SUMO E1/E2 enzymes to prevent APB scaffold nucleation, (2) synthetic-lethal exploitation of replication stress via ATR/CHK1 inhibitors, and (3) immune-microenvironment-targeting strategies that remodel the senescence-associated secretory phenotype (SASP). Collectively, this review elucidates the mechanisms by which ALT regulates cellular senescence and tumorigenesis, offering druggable vulnerabilities and translational strategies for the clinical management of telomerase-negative tumors.
    Citation
    JIA Tong-Xin, XIONG Meng-Jie, HOU Kai-Long, LIU Jia-Hua, ZHANG Hao-Nan, JIA Shu-Ting, LIU Jing.Applications of the Alternative Lengthening of Telomeres Mechanism in Telomerase-Negative Tumors and Senescent Cells[J]..Export: BibTex EndNote
  • Bispecific antibodies, engineered to simultaneously bind two distinct antigens or two epitopes on the same antigen, are now widely utilized in tumor therapy and various other fields. Depending on their mechanisms of action, bispecific antibodies can be designed into diverse structural formats, including IgG-like bispecific antibodies containing an Fc region. The Fc region mediates immune effector functions by interacting with receptors on immune cells or soluble immune components. However, antibodies containing an Fc region have a relatively high molecular weight, which limits their tissue penetration. They also exhibit slow systemic clearance in vivo and possess pharmacokinetic characteristics marked by a long terminal elimination half-life. Symmetric IgG-like bispecific antibodies feature a symmetric structure and are bivalent for each target antigen. During production, since the two heavy chains carrying the Fc region are identical, issues related to chain mispairing do not arise, thereby simplifying the manufacturing and purification processes. Moreover, the pairing of two identical natural Fc chains allows for correct disulfide bond formation, resulting in a more stable structure. Glycosylation of the Fc region remains in its natural state, preserving Fc-mediated functions. However, as the variable regions of the two antigen-binding sites are linked to the same heavy chain, the design must account for potential steric hindrance when the antibody binds both antigens simultaneously. In contrast, asymmetric IgG-like bispecific antibodies consist of two different heavy chains, each carrying antigen-binding domains that recognize distinct antigens or epitopes, offering greater structural design flexibility. Their development, however, requires addressing challenges related to heavy chain and light chain pairing. Strategies to prevent heavy chain mispairing include engineering the spatial configuration of the Fc region, facilitating Fab arm exchange, applying IgG-IgA chain exchange techniques, and introducing charge modifications in the Fc domain. To ensure correct light chain-heavy chain pairing, approaches such as introducing electrostatic interactions or novel disulfide bonds between the chains, swapping the CH1 and CL domains, or replacing the CH1-CL module with a T-cell receptor-derived structure have been employed. Non-IgG-like bispecific antibodies lack an Fc region. They are characterized by their small size and low molecular weight, which confer enhanced tissue penetration, rapid systemic clearance, and high structural versatility. Unlike IgG-based formats, they do not bind Fc receptors or activate the complement system directly. Different bispecific antibodies exert therapeutic effects through distinct mechanisms, which are largely determined by their structural design and target specificity. Currently recognized mechanisms of action include T cell redirection, dual signaling pathway blockade, immune checkpoint inhibition, formation of ternary complexes by binding two molecules, neutralization of soluble ligands, and acting as cofactors to mimic or enhance biological processes. Bispecific antibodies are extensively applied in cancer therapy. Beyond oncology, they are also being developed for the treatment of autoimmune diseases, infectious diseases, hematological disorders, and other conditions. Different structural designs offer unique advantages across therapeutic areas. This article elaborates on the structural designs of various types of bispecific antibodies and reviews their mechanisms of action and applications in therapeutics.
    Citation
    ZHANG Ding, ZHENG Yue-Ting, ZHANG Wei.Structural Design and Application of Bispecific Antibodies[J]..Export: BibTex EndNote
  • Objective Non-coding RNA (ncRNA) plays a crucial regulatory role in various biological processes. Numerous studies have shown that the functions of ncRNAs depend not only on their nucleotide sequences but also closely on their spatial conformations, particularly their secondary structures. Traditional methods for predicting RNA secondary structures often have low accuracy, usually around 50 to 70 percent, and their performance further declines when dealing with complex structures or long sequences. Although deep learning methods have improved prediction performance to some extent, they still face challenges such as high model complexity, difficulty in capturing long-range dependencies, and poor generalization in predicting complex structures. Therefore, it is necessary for RNA secondary structure prediction to develop new methods.Methods This study proposed an end-to-end prediction model RMDfold, which employed a feature extraction strategy combining residual Mamba (resMamba) and dense connections (Dense). The model framework consisted of three modules: one-dimensional (1D) modeling, feature mapping, and two-dimensional (2D) modeling. For the 1D modeling module, the model learned contextual dependencies among nucleotides from RNA sequences, providing the foundation for possible base-pairing; for the feature mapping module, the 1D features were projected into a 2D space to form a constraint matrix that represented potential base-pairing relationships; for the 2D modeling module, the model further learned the pairing patterns between nucleotides, and determined the unique pairing of each nucleotide through pairing constraints, to obtain the final secondary structure. For both 1D and 2D modeling modules, a four-layer Dense block composed of batch normalization, ReLU activation, and convolution was used to extract short-range features; and a dual-branch residual structure resMamba based on a state-space model was used to model long-range dependencies, thereby achieving effective integration of short- and long-range features. To validate the effectiveness of the proposed method, it was compared with Ufold, REDfold, TransUfold, and sincfold on the three public datasets RNAStralign, ArchiveII, and bpRNA-new.Results The proposed RMDfold method demonstrates superior performance compared with existing algorithms in structure prediction, pseudoknot prediction, sequence prediction across varying lengths, and model complexity analysis, while requiring fewer parameters and achieving faster inference. For the structure prediction, the method achieved F1, Matthews correlation coefficient (MCC), precision and recall of 0.973 5, 0.973 1, 0.975 6 and 0.972 3 on RNAStralign, 0.854 3, 0.855 6, 0.874 7 and 0.876 2 on ArchiveII, and 0.382 8, 0.401 5, 0.536 5 and 0.318 7 on bpRNA-new, respectively. For the pseudoknot prediction based on ArchiveII, the model achieved F1, MCC, precision and recall of 0.741 4, 0.743 3, 0.752 5 and 0.739 6. For the sequence prediction across different lengths, RMDfold maintained an accuracy of 0.70 for sequences ranging from 200 to 500 nt. In terms of model complexity, RMDfold required 2.886 7 M parameters and achieves an inference speed of 0.026 0 s.Conclusion RMDfold enables highly accurate prediction of RNA secondary structures. It helps to deeply and comprehensively reveal the central roles of RNA molecules in gene expression regulation, molecular recognition, and catalysis. and also provides important structure support for elucidating disease-related variant mechanisms, designing RNA-targeted drugs, and advancing research in evolution and comparative genomics.
    Citation
    HAO Ze-Zhou, YANG Yan-Ling, ZHOU Liang, YAO Xu-Feng.RMDfold: an End-to-end RNA Secondary Structure Prediction Method Based on Residual Mamba and Dense Connections[J]..Export: BibTex EndNote
  • Viral membrane fusion proteins facilitate the fusion of viral and host cell membranes by undergoing a transition from a prefusion conformation to a post-fusion conformation, thereby enabling the transfer of viral nucleic acids into the cell interior. This transition process is characterized by peptide exposure, membrane insertion, and structural refolding. The prefusion configuration represents an optimal target for vaccine formulation and antiviral pharmacotherapy. However, the metastable nature of the prefusion conformation makes it prone to spontaneous conversion into the stable post-fusion conformation, thereby complicating structural analysis and vaccine design. Investigating the mechanisms of conformational change in these proteins and developing methods to stabilize their prefusion state remain challenging research topics. This review summarizes the structural and functional differences among three classes of membrane fusion proteins: class I proteins, which are predominantly composed of α-helices, form trimers, and rely on receptor binding or low pH to trigger fusion peptide release; class II proteins, which are mainly β-sheet-rich, rearrange from dimers to trimers and activate fusion loops via low pH; and class III proteins, which combine α-helices and β-structures, with mechanisms involving internal fusion loop insertion and membrane remodeling. It is evident that a comprehensive understanding of the mechanisms underlying viral membrane fusion is crucial for developing effective stabilization strategies for the prefusion conformation of these proteins. This paper presents several such methods that have been successfully employed in this endeavor, including: disulfide bond formation to stabilize domain-domain interactions; hydrophobic cavity filling to enhance core stability; proline substitution to restrict structural transitions in hinge regions; and multimer domains stabilizing the trimeric conformation. The stabilization strategies summarized and discussed herein have been validated in studies of multiple viral membrane fusion proteins and further applied in the design of vaccine antigens. Moreover, this paper highlights the potential applications of novel techniques, such as time-resolved cryo-EM, in capturing conformational intermediates and resolving dynamic transition processes. Such stabilization efforts, informed by structural insights, have yielded promising outcomes—for instance, prefusion-stabilized RSV F antigens that elicit potent neutralizing antibodies in clinical trials. Looking ahead, integrating computational modeling, such as AlphaFold predictions, with experimental data will further refine these approaches. Ultimately, these innovations promise to enable structure-guided therapeutics to combat emerging viral threats. This review provides a theoretical foundation for developing stable viral membrane fusion proteins, offering crucial insights for understanding viral membrane fusion mechanisms and advancing next-generation vaccines and antiviral drugs.
    Citation
    XIE Chen-Yi, DONG Xiang-Ge, ZHAN Jiu-Yu, ZHU Hong-Wei, YU Xin, LIU Yang, YU Jia-Yu, ZHANG Xing-Xiao.The Mechanism of Conformational Transition and Stabilisation Strategies of Viral Membrane Fusion Proteins[J]..Export: BibTex EndNote
  • The formation of protein-protein interaction (PPI) networks is a central event in biochemical reactions within organisms. These interactions not only regulate normal physiological functions but are also closely associated with the onset and progression of diseases. PPIs are intricately regulated by proteins, nucleic acids, and their interactions. The complex molecular networks formed between these molecules serve as the foundation for most biochemical reaction events. Moreover, biological information is transmitted through countless molecular interactions within the cellular environment. A wide range of technologies has been developed to study PPIs, among which proximity-dependent biotinylation is a novel technique for labeling proteomes in living cells. This method utilizes engineered biotin ligases to specifically label nearby proteins or RNA molecules, enabling the capture of transient, weak, or stable interactions and facilitating the systematic construction of molecular interaction maps. Through continuous enzyme optimization and refinement, proximity-dependent biotinylation techniques have evolved into diverse systems with improved operational convenience and labeling efficiency. Each proximity-dependent biotinylation technique offers unique advantages: BioID is non-toxic to cells but suffers from low labeling efficiency, requires 18-24 h for labeling, and yields limited biotinylated products. TurboID achieves efficient labeling within 10 min, but its high activity and strong biotin affinity may lead to cytotoxicity. AirID enables low-toxicity labeling under low biotin concentrations but requires several hours to complete. UltraID offers the highest labeling activity with the smallest molecular weight but is prone to over-labeling. APEX provides convenient operation and can resolve protein topology, yet it has concentration-dependent limitations—forming dimers at high concentrations and lacking sensitivity at low concentrations. RNA-BioID is tailored for studying RNA-protein interactions but is limited by non-specific binding. TransitID can capture dynamic protein translocation at the subcellular level, though its temporal resolution still requires improvement. This review systematically summarizes the development, mechanisms, advantages, and disadvantages of proximity-dependent biotinylation techniques such as BioID, TurboID, AirID, UltraID, RNA-BioID, APEX, and TransitID. It also explores their cutting-edge applications in functional regulation and disease research. Proximity-dependent biotinylation techniques are widely used in disease-related studies. In tumor research, they are primarily applied to investigate the transcriptional regulation and chromosomal structural changes of proto-oncogenes and tumor suppressor genes. In the field of neuroscience, they are used to study mechanisms underlying nervous system function and neurological diseases. In viral infection mechanisms, they help elucidate virus-host interaction networks. In immune regulation, they contribute to the study of immune signaling pathways. In stem cell research, they aid in understanding cell differentiation processes. Furthermore, proximity-dependent biotinylation techniques hold promise for integration with spatial biology technologies, enabling more comprehensive and detailed protein studies. These techniques are expected to provide more accurate and efficient tools for life science research and to advance the medical and health fields to a higher level. By comprehensively analyzing the strengths, limitations, and innovative potential of each method, this review also highlights their advantageous applications in molecular interaction studies, aiming to provide methodological guidance and theoretical support for molecular mechanism research in the life sciences.
    Citation
    FANG Tian-Yuan, ZHOU Yu-Yu, ZHANG Chu-Xia, ZHOU Chen-Xuan, CHEN Shi-Yu, LIN Zhi-Cheng, LEI Bin, CHEN Lu-Yi, YING Jia-Qin, CHEN Hui, JIANG Yi, HUANG Yu-Han, LIU Zhi-Tao, LI Li-Ping.Development and Application of Proximal Biotin Labeling Techniques[J]..Export: BibTex EndNote
  • Chinese hamster ovary (CHO) cells are the most established and versatile mammalian expression system for the large-scale production of recombinant therapeutic proteins, owing to their genetic stability, adaptability to serum-free suspension culture, and ability to perform human-like post-translational modifications. More than 70% of biologics approved by the U.S. Food and Drug Administration rely on CHO-based production platforms, underscoring their central role in modern biopharmaceutical manufacturing. Despite these advantages, CHO systems continue to face three persistent bottlenecks that limit their potential for high-yield, reproducible, and cost-efficient production: excessive metabolic burden during high-density culture, heterogeneity of glycosylation patterns, and progressive loss of long-term expression stability. This review provides an integrated analysis of recent advances addressing these challenges and proposes a forward-looking framework for constructing intelligent and sustainable CHO cell factories. In terms of metabolic regulation, excessive lactate and ammonia accumulation disrupts energy balance and reduces recombinant protein synthesis efficiency. Optimization of culture parameters such as temperature, pH, dissolved oxygen, osmolarity, and glucose feeding can effectively alleviate metabolic stress, while supplementation with modulators including sodium butyrate, baicalein, and S-adenosylmethionine promotes specific productivity (qP) by modulating apoptosis and chromatin structure. Furthermore, genetic engineering strategies—such as overexpression of MPC1/2, HSP27, and SIRT6 or knockout of Bax, Apaf1, and IGF-1R—have demonstrated significant improvements in cell viability and product yield. The combination of multi-omics metabolic modeling with artificial intelligence (AI)-based prediction offers new opportunities for building self-regulating CHO systems capable of dynamic adaptation to environmental stress. Regarding glycosylation uniformity, which determines therapeutic efficacy and immunogenicity, gene editing-based glycoengineering (e.g., FUT8 knockdown or ST6Gal1 overexpression) has enabled the humanization of CHO glycan profiles, minimizing non-human sugar residues and enhancing drug stability. Process-level strategies such as galactose or manganese co-feeding and fine control of temperature or osmolarity further allow rational regulation of glycosyltransferase activity. Additionally, in vitro chemoenzymatic remodeling provides a complementary route to construct human-type glycans with defined structures, though industrial applications remain constrained by cost and scalability. The integration of model-driven process design and AI feedback control is expected to enable real-time prediction and correction of glycosylation deviations, ensuring batch-to-batch consistency in continuous biomanufacturing. Long-term expression stability, another critical challenge, is often impaired by promoter silencing, chromatin condensation, and random genomic integration. Molecular optimization—such as the use of improved promoters (CMV, EF-1α, or CHO endogenous promoters), Kozak and signal peptide refinement, and incorporation of chromatin-opening elements (UCOE, MAR, STAR)—helps maintain durable transcriptional activity, while site-specific integration systems including Cre/loxP, Flp/FRT, φC31, and CRISPR/Cas9 can enable single-copy, position-independent gene insertion at genomic safe-harbor loci, ensuring stable, predictable expression. Collectively, this review highlights a paradigm shift in CHO system optimization driven by the convergence of genome editing, synthetic biology, and artificial intelligence. The transition from empirical optimization to rational, data-driven design will facilitate the development of programmable CHO platforms capable of autonomous regulation of metabolic flux, glycosylation fidelity, and transcriptional activity. Such intelligent cell factories are expected to accelerate the transformation from laboratory-scale research to industrial-scale, high-consistency, and economically sustainable biopharmaceutical manufacturing, thereby supporting the next generation of efficient and customizable biologics manufacturing.
    Citation
    ZHANG Rui-Ming, LI Meng-Lin, ZHU Hong-Wei, ZHANG Xin-Xiao.Strategic Optimization of CHO Cell Expression Platforms for Biopharmaceutical Manufacturing[J]..Export: BibTex EndNote
  • Post-translational modification (PTM) of proteins refers to the covalent addition of functional groups to amino acid residues or structural alterations in proteins during or after translation, primarily mediated by enzymatic reactions and secondarily by non-enzymatic chemical processes. PTM crosstalk denotes interactions between distinct modification sites or different types of modifications on a single protein, which regulate protein functions through synergistic, antagonistic, or cascading mechanisms. Lactylation, phosphorylation, and acetylation are three pivotal types of protein PTMs, involving the covalent attachment of lactic acid, phosphate, and acetyl groups to specific amino acid residues, respectively. These reversible modifications are dynamically regulated by cellular metabolic status and signaling pathways. Phosphorylation primarily facilitates rapid signal transduction; acetylation broadly regulates metabolism and gene expression; and lactylation is closely associated with high-lactate microenvironments and metabolic stress. Through competitive binding at identical or adjacent sites, reciprocal modulation of metabolite levels, and cross-regulation of signaling pathways, these three modifications form an intricate crosstalk network that coordinately regulates cellular adaptive responses to internal and external environmental changes. As a physiological stimulus with broad effects on bodily functions, exercise induces a series of changes in intracellular metabolism and signal transduction, thereby influencing PTMs and their crosstalk. On one hand, exercise activates multiple interconnected cellular systems, including energy metabolism, signal transduction, and molecular interaction networks. Within the energy metabolism system, exercise alters the pattern of cellular ATP production and utilizes metabolic intermediates as signaling molecules to directly or indirectly modulate the activity of enzymes involved in these three modifications. In the signal transduction system, exercise activates pathways such as AMP-activated protein kinase (AMPK) and mitogen-activated protein kinase (MAPK), which precisely regulate the activity and subcellular localization of modification-related enzymes via phosphorylation cascades. In the molecular interaction system, exercise promotes protein-protein and protein-metabolite interactions, thereby remodeling the regulatory network of PTMs. On the other hand, exercise facilitates crosstalk among lactylation, phosphorylation, and acetylation through a multi-level progressive regulatory model: “metabolic initiation → signal transduction → molecular interaction”. At the metabolic level, alterations in metabolites provide the initial driving force for crosstalk; signaling pathways amplify these signals and precisely modulate the direction of crosstalk through cascade reactions; and molecular interactions further integrate signals to establish a refined regulatory network. Ultimately, this multi-system and multi-level crosstalk enables precise regulation of cell proliferation, differentiation, and apoptosis, thereby mediating cellular adaptation to exercise and playing a central role in enhancing exercise capacity and improving metabolic health. This article systematically examines how exercise influences crosstalk among these three key PTMs—lactylation, phosphorylation, and acetylation—and the underlying mechanisms, including the regulation of metabolite levels, modification-related enzyme activity, cellular signaling pathways, metabolic homeostasis, and gene expression. This work provides a novel perspective for gaining deeper insights into how exercise regulates physiological functions.
    Citation
    LI Ting-Ting, LIU Yu, LI Hong, WANG Shi-Da, ZHANG Hai-Feng.The Effects and Mechanisms of Exercise on The Crosstalk Among Post-translational Modifications of Proteins[J]..Export: BibTex EndNote
  • Obesity has become a major global public health concern, affecting more than one billion individuals worldwide. As a low-grade chronic inflammatory condition, obesity is closely associated with cardiometabolic disorders and gut microbial dysbiosis. Diet-based interventions are recognized as one of the safest and most effective strategies for long-term weight management. Increasing evidence indicates that specific dietary patterns can modulate gut microbiota (GM) composition and metabolic function. However, comparative evidence regarding the effects of different dietary strategies remains limited and inconsistent. This systematic review and meta-analysis comprehensively evaluated the effects of the very-low-calorie ketogenic diet (VLCKD), Mediterranean diet (MD), and intermittent fasting (IF) on gut microbiota in obese populations. Systematic searches of PubMed, EBSCOhost, Cochrane, and Web of Science were conducted up to September 2025. Meta-analyses using R software assessed changes in microbial diversity and characteristic taxa abundance, with subgroup analyses by body mass index (BMI), age, and intervention duration. A total of 42 studies were included. Random-effects meta-analysis revealed that VLCKD significantly increased the Shannon index, observed OTUs, and Faith"s phylogenetic diversity (PD), promoted Akkermansia abundance and the Firmicutes/Bacteroidetes (F/B) ratio, but reduced Bifidobacterium abundance, indicating a bidirectional regulatory effect on gut microbial structure. MD significantly increased the Shannon index as well as the abundance of Akkermansia, Bifidobacterium, and Bacteroidetes, while decreasing Firmicutes abundance and the F/B ratio, suggesting a balanced and sustained improvement in gut microbial composition. In contrast, IF significantly decreased the PD index while increasing Akkermansia and reducing Firmicutes, reflecting partial structural optimization but limited enhancement of phylogenetic diversity; long-term interventions were associated with a decline in Shannon diversity, indicating limited stability. Subgroup analyses revealed distinct moderator effects. Under VLCKD, improvements in microbial diversity were more pronounced among individuals with BMI≤30 kg/m2 and those aged >30 years, and meta-regression confirmed that the magnitude of diversity gains increased with age. Regarding BMI, increases in Akkermansia abundance were most evident in individuals with BMI 30-35 kg/m2, whereas Bifidobacterium abundance significantly decreased in the same range, suggesting a threshold-dependent microbial response to adiposity. With respect to age, both Akkermansia (increase) and Bifidobacterium (decrease) exhibited significant changes in individuals aged >40 years. In terms of intervention duration, Akkermansia increased significantly within 6 weeks, while Bifidobacterium decreased within 12 weeks. For MD, increases in Shannon diversity were consistently observed across all BMI, age, and duration subgroups; notably, Akkermansia abundance increased significantly among participants with BMI>30 kg/m2, aged 30-50 years, and during interventions ≤6 months, while Bifidobacterium abundance rose markedly in participants with BMI≤30 kg/m2, aged 40-50 years, and during interventions of 6-12 months. Under IF, Shannon diversity increased significantly in individuals with BMI≤30 kg/m2 and aged >40 years but declined when the intervention exceeded 4 weeks, suggesting reduced long-term stability. In conclusion, VLCKD, MD, and IF all modulate gut microbiota in obesity but differ in magnitude, direction, and durability. VLCKD exerts strong yet dual effects—enhancing diversity while reducing beneficial taxa; MD shows stable, sustained modulation; whereas IF offers selective improvements but lowers phylogenetic diversity with limited persistence. Future studies should conduct large, multicenter randomized controlled trials to determine optimal intervention duration, confirm the moderating roles of age and BMI, and develop personalized microbiota-based dietary strategies for obesity management and gut health improvement.
    Citation
    WANG Shun, BAO Zi-Han, ZHAO Meng-Qi, LI Zi-Yang, WANG Xun-Ling, LIU Feng-Hu.Impact of Dietary Pattern Interventions on Gut Microbiota in Obesity: a Systematic Review and Meta-analysis[J]..Export: BibTex EndNote
  • The comorbidity of sarcopenia and cognitive impairment constitutes a degenerative syndrome that progresses significantly with age. It has emerged as a critical global health challenge, contributing to functional disability, reduced quality of life, and increased pressure on public healthcare systems. This comorbidity is characterized by a synergistic decline in both physical and cognitive capabilities, manifesting as reduced skeletal muscle mass, diminished muscle strength, impaired physical function, and progressive deterioration in cognitive domains such as memory, executive function, and information processing speed. This dual degeneration not only creates a vicious cycle where each condition exacerbates the other but also substantially increases the risk of falls, fractures, hospitalization, and mortality among older adults. Against the backdrop of rapid global population aging, the prevalence of this comorbidity is anticipated to rise further without effective interventions. Consequently, investigating its underlying mechanisms and developing preventive and therapeutic strategies hold substantial clinical and public health significance. Current evidence indicates that the pathogenesis involves multi-system and multi-level pathophysiological processes, with chronic inflammation, mitochondrial dysfunction, and gut microbiota dysbiosis, identified as three core interacting mechanisms. Age-related chronic low-grade inflammation, termed inflammaging, arises from the senescence-associated secretory phenotype (SASP) and persistent immune cell activation. This inflammatory state inhibits the intramuscular IGF-1/Akt/mTOR anabolic pathway through proinflammatory cytokines (e.g., IL-6, TNF-α), while simultaneously activating protein degradation systems including the ubiquitin-proteasome system (UPS) and autophagy-lysosomal pathway (ALP), ultimately leading to accelerated protein breakdown and muscle atrophy. These circulating inflammatory factors can also compromise blood-brain barrier integrity, activate microglia, trigger neuroinflammation, and consequently damage synaptic structures and neuronal function, thereby accelerating cognitive decline in this comorbidity. Mitochondrial dysfunction presents as impaired oxidative phosphorylation efficiency, excessive reactive oxygen species (ROS) production, and dysregulated mitochondrial quality control. This not only results in inadequate cellular energy supply but also enables mitochondrial-derived factors (e.g., extracellular mtDNA) to activate innate immune pathways such as cGAS-STING, propagating stress signals and amplifying tissue damage in both muscle and brain. Additionally, gut microbiota dysbiosis impairs intestinal barrier function, increases lipopolysaccharide (LPS) translocation into circulation, and reduces short-chain fatty acid (SCFA) production. These changes induce systemic inflammation and metabolic disturbances that further impact muscle metabolism and promote pathological protein accumulation in the brain, thereby establishing a gut-brain-muscle axis that exacerbates the progression of this comorbidity. Exerkines represent a class of biologically active signaling molecules—including cytokines, peptides, metabolites, and exosomes—secreted by various tissues in response to exercise. These exerkines mediate systemic adaptations and protective effects through endocrine and paracrine actions on target organs. Key exerkines such as IL-6, Irisin, brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1), fibroblast growth factor 21 (FGF-21), lactate, and cathepsin B (CTSB) play central roles in coordinately ameliorating the comorbidity of sarcopenia and cognitive impairment. The beneficial effects of these exerkines are mediated through multiple mechanisms including inflammation modulation, energy metabolism remodeling, neuroprotection, and enhanced neuroplasticity. As a non-pharmacological intervention, exercise effectively stimulates the production and release of exerkines, thereby targeting the comorbidity through multiple pathways. Aerobic exercise elevates lactate levels and activates the Sirt1/PGC-1α pathway, improving cerebral metabolism and cognitive function. Resistance training significantly upregulates IGF-1, Irisin, and CTSB expression, enhancing muscle anabolism and hippocampal function. Other modalities like high-intensity interval training (HIIT) and traditional practices also help modulate inflammatory status and optimize the neurotrophic environment through the action of various exerkines. Different exercise types work synergistically by engaging distinct signaling pathways and exerkine combinations, collectively alleviating chronic inflammation, correcting mitochondrial dysfunction, and optimizing gut microecology to achieve concurrent musculoskeletal and cognitive protection against this comorbidity. Synthesizing current evidence, this review emphasizes the necessity of transcending a single-organ perspective by recognizing muscle and brain as an integrated functional unit, with exerkines playing a pivotal role in the muscle-brain axis. The field nevertheless faces several challenges: the secretion dynamics of exerkines during aging remain unclear, mechanisms underlying individual differences in exercise response require elucidation, and the compensatory and imbalance characteristics of exercise-induced exerkine networks across disease stages need further characterization. Future research should employ large-sample cohorts and randomized controlled trials integrated with multi-omics technologies to establish personalized exercise interventions based on exerkine profiling for managing this comorbidity. Parallel efforts should focus on developing quantifiable efficacy assessment systems to provide robust theoretical foundation and practical guidance for precise management of the comorbidity of sarcopenia and cognitive impairment and the promotion of healthy aging.
    Citation
    XIA Jun-Mei, DENG Qi, HAO Hong-Tao, LIANG Ji-Ling.The Mechanism of Exerkines on The Comorbidity of Sarcopenia and Cognitive Impairment[J]..Export: BibTex EndNote
  • Objective To address the challenge of identifying unknown animal-derived components in the fields of forensic evidence and food safety, this study developed and validated a sensitive and efficient multiplex polymerase chain reaction-capillary electrophoresis (PCR-CE) detection system capable of simultaneously identifying DNA from 13 species—cattle (Bos taurus), mouse (Mus musculus), dog (Canis lupus familiaris), rat (Rattus norvegicus), pig (Sus scrofa), Chinese hamster (Cricetulus griseus), cat (Felis catus), horse (Equus caballus), human (Homo sapiens), chicken (Gallus gallus), duck (Anas platyrhynchos), donkey (Equus asinus), and sheep (Ovis aries)—within a single-tube reaction.Methods Species-specific primers were meticulously designed targeting hypervariable regions of the mitochondrial DNA (including Cytb, COI, 16S rRNA, and ND2 genes), with primer specificity rigorously verified in silico using BLAST analysis against non-target species. A multiplex PCR system was constructed, and critical reaction parameters, including primer concentrations and annealing temperature, were systematically optimized through gradient experiments to ensure balanced amplification of all 13 targets without non-specific products or primer-dimer formation. Amplification products were subsequently separated and detected based on their characteristic fragment lengths and distinct fluorescent labels (6-FAM, HEX) using a capillary electrophoresis platform. The established multiplex PCR-CE system was comprehensively evaluated across several parameters: specificity was tested against 17 species (13 target species and 4 non-target species); sensitivity was determined using serial dilutions of mixed DNA templates; the ability to detect adulteration was assessed using simulated mixed meat samples with known adulteration percentages (10% to 0.1%); and practical applicability was investigated by analyzing 27 commercially available meat products and authentic casework samples from a fatal dog attack incident.Results The optimized multiplex PCR-CE system successfully demonstrated specific and simultaneous amplification for all 13 target species, with zero cross-reactivity observed with 4 non-target species. The system exhibited high sensitivity, with detection limits ranging from 0.05 ng to 0.001 ng of DNA template depending on the species; ten species were detectable at the 0.001 ng level. In simulated adulteration studies, the system reliably detected duck DNA in sheep meat at 0.5%, pork in beef at 0.5%, and horse meat in donkey meat at a remarkably low level of 0.1%. Analysis of commercial meat products revealed an 18.52% (5/27) mislabeling rate. These findings were consistent with validation tests using national and industry standard methods. Furthermore, the system effectively identified human and dog DNA from real forensic case evidence (clothing fragments from bite marks), and even detected trace pig DNA, suggesting the dog"s prior pork consumption.Conclusion This study successfully established a highly specific, sensitive, and practical multiplex PCR-CE system for the simultaneous identification of 13 species. By combining the power of multiplex PCR with capillary electrophoresis, our approach delivers a significant advantage in terms of throughput, resolution, and automation over traditional gel-based methods. The system"s proven effectiveness in detecting low-level adulteration in complex mixtures and processed food products, along with its successful application to genuine forensic specimens, underscores its substantial value and broad potential for routine use in forensic laboratories for evidence analysis and in regulatory settings for ensuring food authenticity and safety monitoring.
    Citation
    GUO Run-Ze, HU Sheng, CHEN Li-Ping, ZHANG Ying, HE Yong-Feng, SONG Zhen, YANG Rui-Qin, SUN Qi-Fan.Development and Validation of a Multiplex PCR-capillary Electrophoresis System for Identification of Frequently Encountered Species[J]..Export: BibTex EndNote
  • Social interaction is central to the development of human cognition and behavior. Studying the neural mechanisms of social interaction helps reveal the neurobiological basis of social functions, such as group cooperation and knowledge transfer. In recent years, social neuroscience research has adopted hyperscanning technology and brain-to-brain coupling (BBC) measurements to reveal the group neural dynamics mechanisms under social interactions. Existing studies have primarily focused on three social contexts highly relevant to social interaction, namely interpersonal communication, task collaboration, and teacher-student instruction. However, the driving factors of BBC across these contexts and their interaction patterns under naturalistic paradigms have not yet been systematically analyzed within a unified framework, and the underlying driving mechanisms remain unclear. To address this limitation, the present review focused on three social contexts with increasing ecological validity in social interaction. It systematically examines the exogenous and endogenous drivers of BBC across these contexts and reveals commonalities and differences across contexts. Exogenous factors provide external conditions and spatiotemporal framework for interaction through sensory input and behavioral patterns. These external conditions induce BBC by guiding individuals to focus on the same target within the same time window, thereby invoking shared attention. However, exogenous drivers can only ensure surface alignment of interactions. Without the support of endogenous drivers, brain-brain coupling is difficult to maintain or deepen. Endogenous factors determine the depth and continuity of interactions through high-level social cognitive processing. Specifically, social closeness enhances trust and empathy between interacting partners, promoting interpersonal multimodal information integration and emotional empathy. Shared attention is the key link for individuals to move from behavioral alignment to initial coupling at the neural level. Shared intentionality led individuals to converge on goals and strategies, forming cognitive predictions during the cooperation process. Shared understanding ensures that individuals can perform high-level cognitive processing based on a common knowledge framework. The above-mentioned endogenous driving factors enhance BBC by engaging higher-order cognitive regions such as the prefrontal cortex, temporoparietal junction, and default mode network. This engagement enables the interaction to shift from transient attention coupling to stable intention alignment and cognitive sharing. Therefore, the formation of BBC can be viewed as a process that evolves "from external to internal, from weak to strong". Exogenous driving factors initiate neural alignment through shared attention. Endogenous driving factors then strengthen BBC via shared understanding and shared intentionality. Together, these processes support the construction of group-level shared cognition. Finally, this article summarized the current challenges in research on the driving mechanisms of BBC and provided an outlook for future development. First, it is necessary to theoretically establish a hierarchical model of brain-brain coupling based on a hierarchy of cognitive complexity, systematically distinguishing between the characteristics of BBC driven by low-level processes and higher-level interpersonal shared cognitive mechanisms. Second, at the methodological and technical level, future development of multimodal hyperscanning systems such as EEG-fNIRS and closed-loop hyper-transcranial alternating current stimulation could comprehensively analyze the dynamic evolution of BBC related to shared cognition in the temporal, spatial, and frequency domains. In summary, this article constructed a theoretical framework for the driving mechanisms of BBC across social contexts, hoping to provide a methodological basis for controlling the driving factors of naturalistic paradigms in social neuroscience research.
    Citation
    WANG Zheng-Yi, SHU Lei-Jin, YU Hai-Qing, CHEN Yuan-Fang, XU Min-Peng, JUNG Tzyy-Ping, MING Dong.Multi-contextual Driving Mechanisms of Brain-to-brain Coupling in Social Interaction[J]..Export: BibTex EndNote
  • This review synthesizes recent advances in prolamin-based multicomponent nanocarriers, with a focus on their physicochemical properties, modification strategies, and potential applications in functional foods, biomedicine, and sustainable agriculture. The abundance of hydrophobic amino acid residues in prolamins facilitates spontaneous self-assembly into nanoparticles, making them promising carriers for poorly water-soluble bioactive compounds such as curcumin and resveratrol. However, native prolamin nanoparticles suffer from limitations including poor colloidal stability, tendency to aggregate under processing or physiological conditions (e.g., pH, ionic strength, enzymatic degradation), and limited functional diversity. To address these drawbacks, extensive research has been devoted to modification strategies aimed at enhancing stability, structural integrity, and cargo protection. Polysaccharide modification enables the formation of stable core-shell structures through electrostatic interactions, hydrogen bonding, and steric hindrance. Coatings with pectin, chitosan, or alginate improve stability across a broad range of pH values and ionic strengths, enhance resistance to gastric digestion, and enable sustained release in the intestine, thereby improving bioavailability. Polyphenol modification introduces hydrogen bonding, hydrophobic interactions, and occasionally covalent cross-linking, which modify nanoparticle structure and surface properties. These composites exhibit improved hydrophilicity, colloidal stability, and resistance to oxidative or UV-induced degradation, along with intrinsic antioxidant activity. Lipid modification leverages hydrophobic interactions with oils or fatty acids to form composite nanoparticles or Pickering emulsions. This approach increases the loading capacity for hydrophobic compounds, creates a protective barrier, and enhances oral bioavailability by promoting emulsification and intestinal absorption. Additional strategies include the incorporation of auxiliary proteins (e.g., casein, whey protein) to improve stability and emulsifying capacity, as well as the use of inorganic nanomaterials (e.g., SiO2, AuNPs) to impart mechanical reinforcement, antibacterial properties, and stimuli-responsive functions. Genetic engineering further allows molecular-level tailoring of amino acid sequences to fine-tune hydrophobicity, amphiphilicity, and self-assembly behavior. These engineered nanocarriers exhibit advanced functionalities. They enable sustained and stimuli-responsive release triggered by pH, redox potential, enzymes, temperature, or light, facilitating on-demand delivery that maximizes efficacy while minimizing off-target effects. Targeting can be achieved passively through the enhanced permeability and retention (EPR) effect, or actively via conjugation with ligands, antibodies, or peptides that recognize specific receptors. The applications of these systems are broad. In functional foods and nutraceuticals, prolamin-based carriers improve the stability, bioavailability, and controlled release of sensitive bioactive ingredients, supporting personalized nutrition. In biomedicine, they enhance oral drug delivery, enable targeted cancer therapy with reduced systemic toxicity, and serve as scaffolds for tissue engineering. In agriculture, they facilitate the controlled release of pesticides, fertilizers, and growth regulators, helping to reduce environmental contamination and promote sustainable practices; they are also being explored for smart food packaging applications. Despite significant progress, challenges remain in clinical and industrial translation. There is an urgent need for standardized characterization methods, comprehensive in vivo safety and efficacy evaluations, and scalable, regulation-compliant manufacturing processes. Future research should adopt rational design principles to develop multi-stimuli-responsive and sustainable systems. The integration of artificial intelligence and data-driven approaches may further accelerate the development of personalized theranostic platforms and co-delivery systems. Continued innovation is expected to solidify the role of prolamin-based nanocarriers in advancing global health and sustainable development.
    Citation
    HUANG Yun-Jie, Qi Li-Li, MEI Han-Fang, WANG Meng-Ting, YU Yong, WANG Jin-Bo.From Self-assembly to Smart Delivery: Construction Strategies and Frontier Applications of Prolamin-based Multicomponent Complex Nanocarriers[J]..Export: BibTex EndNote
  • Alzheimer’s disease (AD) is a common chronic neurodegenerative disorder of the central nervous system characterized by progressive impairments in memory, cognition, and behavior, eventually leading to severe dementia and loss of self-care ability. Despite decades of investigation, the precise molecular mechanisms underlying AD remain incompletely understood, and effective disease-modifying treatments are still lacking. The traditional pathological hallmarks of AD including amyloid β-protein (Aβ) plaques and neurofibrillary tangles (NFTs) composed of hyperphosphorylated Tau fail to account for the complex biochemical and cellular alterations observed in AD brains. Ferroptosis, a distinct iron-dependent form of non-apoptotic programmed cell death, is increasingly recognized as a contributor to AD pathogenesis. Ferroptosis is driven by excessive accumulation of lipid peroxides and reactive oxygen species (ROS), leading to oxidative destruction of cellular membranes. Unlike apoptosis or necrosis, ferroptosis is morphologically characterized by shrunken mitochondria with condensed membrane densities and biochemically defined by the loss of glutathione peroxidase 4 (GPX4) activity. Disruption of iron homeostasis, a central hallmark of ferroptosis, triggers a cascade that inhibits the cystine/glutamate antiporter (System Xc-), suppresses glutathione (GSH) synthesis, and impairs GPX4-mediated detoxification of lipid peroxides, leading to uncontrolled lipid peroxidation and oxidative stress that ultimately trigger ferroptotic cell death. This iron-driven cell death exhibits distinct morphological and biochemical characteristics compared with other forms of cell death. Ferroptosis contributes to AD pathogenesis through multiple mechanisms and is closely associated with disease onset and progression. Iron overload can affect early amyloid precursor protein processing, accelerate Aβ production and plaque deposition, reduce Tau protein solubility, and promote Tau hyperphosphorylation and aggregation into NFTs. Therapeutic strategies targeting ferroptosis—such as iron chelation with deferoxamine to reduce labile iron levels and inhibit Fenton reaction-driven oxidative damage; supplementation with antioxidants such as α-tocopherol or α-lipoic acid to neutralize reactive oxygen species (ROS) and scavenge lipid radicals; and administration of selenium or activators of the Nrf2-SLC7A11-GPX4 axis and the SIRT1/Nrf2 signaling pathway to restore glutathione-GPX4 function—can effectively block lipid peroxidation and suppress iron-dependent cell death. By modulating iron metabolism, enhancing antioxidant defenses, and inhibiting lipid peroxidation, these approaches hold promise for mitigating ferroptosis-related neuronal injury. These interventions collectively aim to modulate iron metabolism, strengthen antioxidant defenses, and suppress lipid peroxidation, thereby mitigating neuronal injury and delaying cognitive deterioration. Ferroptosis represents a pivotal intersection of iron metabolism, oxidative stress, and neurodegeneration in AD. Exploring ferroptotic mechanisms not only deepens our understanding of AD pathophysiology but also opens new avenues for therapeutic intervention. This review aims to comprehensively summarize the molecular basis of ferroptosis, elucidate its pathological roles in AD, and propose ferroptosis-centered therapeutic strategies, thereby providing a theoretical framework for future research and drug development in AD.
    Citation
    LEI Bin, YING Jia-Qin, CHEN Shi-Yu, LIN Zhi-Cheng, LI Wan-Yi, LIU Zhi-Tao, HUANG Yu-Han, YE Zhi-Tao, CHEN Lu-Yi, ZHOU Chen-Xuan, JIANG Yi, CHEN Hui, YANG ZI-YU, LI Li-Ping.Ferroptosis in Alzheimer’s Disease: Potential Mechanisms and Intervention Strategies[J]..Export: BibTex EndNote
  • Microorganisms, as one of the Earth’s most abundant genetic resources, demonstrate tremendous application potential in fields such as medicine, energy, and environmental protection. However, natural microorganisms often suffer from poor stability and low catalytic efficiency. The emergence of microorganism-nanomaterial hybrid systems offers novel strategies to overcome these limitations. These systems integrate nanomaterials with microorganisms or their components (e.g., cell membranes, metabolites, or biomacromolecules) through methods such as biomineralization, electrostatic assembly, surface modification, and genetic engineering. This enables programmable design from the nanoscale to the macroscale, demonstrating broad application prospects and attracting extensive research interest. First, microbial-nanomaterial hybrid systems are classified based on the types of nanomaterials (organic, inorganic, organic-inorganic) and microorganisms (bacteria, fungi, viruses, algae, probiotics). Both types of systems leverage the unique catalytic selectivity of microorganisms and the diverse physicochemical properties of nanomaterials to achieve multidimensional synergy. Their synergistic mechanisms involve both the biochemical processes of microorganisms and the surface/interface reactions of nanomaterials, representing a multidisciplinary achievement spanning microbial interface engineering, biomimetic catalysis, controllable nanomaterial fabrication, and interfacial transport and reaction processes. Next, the application progress in biomedical fields (such as anti-infection, intestinal diseases, and cancer therapy) and energy conversion (e.g., light-driven hybrid systems for proton reduction to hydrogen, CO2 reduction and conversion, and nitrogen fixation) is elaborated in detail, highlighting their significant advantages in functional integration and synergistic performance. Microorganism-nanomaterial hybrid systems combine the specific recognition and precise metabolic capabilities of microorganisms with the catalytic, drug-delivery, and optoelectronic functions of nanomaterials, enabling the construction of various multifunctional synergistic platforms for catalysis, diagnosis, and therapy. These advances have greatly promoted development in nanomedicine, energy, and environmental applications. In medical contexts, such systems utilize the natural chemotaxis of microorganisms for precise targeting, achieve controlled drug release through environmentally responsive delivery and metabolic regulation, and enhance therapeutic efficacy via combined chemical-biological treatments and immune modulation. Improved biosafety can be achieved through attenuated microbial designs and nanomaterial coatings, offering diverse strategies for the precise treatment of various diseases. In the energy sector, the excellent light-harvesting properties of semiconductor materials and the precise catalytic capabilities of biological systems have been integrated to successfully construct light-driven biocatalytic systems, significantly improving light utilization efficiency. Finally, this review discusses the key challenges facing the practical application of these systems. Nanomaterials may exert toxic effects on microorganisms, impairing their activity and raising environmental safety concerns. The potential release of engineered nanomaterials into ecosystems necessitates careful risk assessment and long-term monitoring. In real-world environments, microbial functions are easily compromised, nanostructures are prone to damage, and reactive oxygen species (ROS) tend to accumulate, resulting in insufficient system stability. Stringent culture conditions, costly raw materials, and significant batch-to-batch variability hinder large-scale production and commercialization. The synergistic mechanisms between microorganisms and nanomaterials are not yet fully understood, particularly regarding molecular-level interactions and long-term compatibility. In medical applications, off-target risks persist due to unpredictable microbial colonization and immune responses, while environmental applications lack sufficient selective recognition capabilities, indicating a need for improved targeting and specificity. Furthermore, interdisciplinary barriers between biology, materials science, and engineering complicate collaborative innovation, and the absence of well-established standards for evaluation, regulation, and scalability also constrains further development. Future efforts should focus on enhancing biocompatibility, optimizing fabrication processes, and establishing comprehensive safety and performance standards to accelerate the transition of these promising systems from laboratory research to real-world applications.
    Citation
    CHEN Ren-Ju, LUO Bang-Lan, QUAN Chun-Shan, LI Chun-Bin, LIN Feng, ZHANG Yan-Mei.Microbial-Nanomaterial Hybrid Systems[J]..Export: BibTex EndNote
  • Objective With the continuous evolution of severe acute respiratory syndromes-coronary virus 2 (SARS-CoV-2) Omicron subvariants, particularly the emergence of BA.2.86 and its descendant JN.1, the efficacy of current neutralizing antibodies has faced substantial challenges. The JN.1 variant, noted for its pronounced immune evasion capacity, has rapidly become the globally dominant strain. Elucidating its escape mechanisms is therefore essential to guide the development of next-generation broad-spectrum vaccines and neutralizing antibody therapeutics. This study aimed to investigate the immune evasion mechanisms of JN.1 against broadly neutralizing antibodies, focusing on the effects of key receptor-binding domain (RBD) mutations on antibody binding and neutralization, thereby providing theoretical support for countering ongoing viral evolution.Methods We employed a multidisciplinary approach to systematically assess the binding and neutralizing activities of three broad-spectrum neutralizing antibodies (XGv074, XGv302, and XGv303) against BA.2.86 and JN.1. Binding affinities (KD values) of antibodies to variant RBDs were determined using bio-layer interferometry (BLI). Cryo-electron microscopy (cryo-EM) was used to resolve the structure of the BA.2.86 Spike trimer in complex with antibody antigen-binding fragments (Fabs), achieving a resolution of 3.47 ? for the BA.2.86 S-trimer bound to XGv302. Molecular dynamics simulations and binding free-energy decomposition were conducted to quantify the contributions of key mutations at the antibody-RBD interface. Additionally, sequence alignment and structural modeling were performed to evaluate the role of conformational flexibility in the antibody heavy-chain complementarity-determining region 3 (HCDR3) in mediating tolerance to mutations.Results Experimental data showed that XGv074, XGv302, and XGv303 retained neutralizing activity against BA.2.86 but exhibited markedly reduced binding to JN.1, with only XGv074 maintaining weak neutralization (IC50=2.3 mg/L). Cryo-EM structures revealed that all three antibodies targeted the RBD tip, overlapping with the ACE2-binding region. The JN.1-specific L455S mutation disrupted the hydrophobic interaction network between XGv302 and the RBD (involving key residues such as Y421 and L455), resulting in complete loss of neutralization. Binding free-energy decomposition further identified L455 and Y421 as energetic hotspots (ΔG<-3 kcal/mol), with the L455S mutation directly impairing antibody binding. XGv074, owing to greater conformational flexibility in its HCDR3 region, partially tolerated the mutation and retained weak binding. Molecular dynamics simulations showed that the L455S mutation not only eliminated the energetic contribution of this residue but also caused a concurrent decrease in binding free energy of neighboring residues, thereby reducing overall interface stability.Conclusion The JN.1 variant escapes broad-spectrum neutralizing antibodies primarily through the L455S mutation in the RBD, which disrupts energetic hotspots and remodels the antibody-binding interface. Antibody conformational flexibility enhances adaptability to such mutations, providing new insights for broad-spectrum antibody design. These findings highlight the critical roles of epitope energy distribution and antibody flexibility in maintaining neutralization breadth, offering essential guidance for the rational design of next-generation vaccines and antibody therapeutics: specifically targeting conserved energetic hotspots while enhancing CDR flexibility to counter immune evasion driven by viral evolution.
    Citation
    XIE Jia-Wen, LIU Tian-Ci, GUO Meng-Tian, FENG Lu-Lu, SUN Ming-Chen, LIU Pan, ZHU Qian-Hui.Mechanisms of Immune Evasion by The SARS-CoV-2 JN.1 Variant Against Broadly Neutralizing Antibodies[J]..Export: BibTex EndNote
  • Objective This study proposes a fatigue detection method for police extreme training based on electrical impedance imaging technology to prevent muscle damage caused by overstrain during intense physical training.Methods First, based on the mechanism of human anaerobic exercise, lactic acid was identified as a key indicator of muscle fatigue, demonstrating that measuring muscle lactic acid effectively reflects localized fatigue. Second, a numerical simulation model of the human calf was established, and the internal tissue structure of the calf was analyzed to determine the stages of lactic acid diffusion and change. Then, the reconstruction performance of electrical impedance tomography (EIT) in visualizing lactic acid diffusion was compared under three different regularization algorithms, and the most suitable regularization method for subsequent experiments was selected. Finally, a controlled experiment simulating lactate diffusion was conducted to verify the imaging capability of the TK-Noser regularization algorithm in complex imaging fields.Results Simulation results indicate that both the TK-Noser and TV regularization algorithms achieve superior imaging performance, effectively suppressing artifacts in the visualization of lactic acid diffusion inside muscle tissue. The average ICC/RMSE values reached 0.754/0.303 and 0.772/0.320, respectively, while the average SSIM/PSNR values were 0.677/61 dB and 0.488/60 dB, respectively. In the lactate diffusion experiment, the average ICC/SSIM of the EIT reconstruction results based on the TK-Noser regularization algorithm reached 0.701 and 0.572, respectively. Additionally, compared with the TV regularization algorithm, the TK-Noser algorithm better preserved the shape and structural integrity of the imaging target, with an SSIM value 21.2% higher than that of the TV regularization results. This enhancement ensures the stability of the experimental results and significantly improves the capability of electrical impedance imaging technology in monitoring lactate diffusion within complex fields.Conclusion The proposed method offers real-time convenience and non-invasiveness, making it a promising approach for dynamic monitoring of muscle lactate levels in police officers during extreme physical training.
    Citation
    LIU Tao, SHI Shu-Sheng, LIU Jun-Feng, LIU Kai, YAO Jia-Feng.A Muscle Fatigue Assessment Method of Electrical Impedance Tomography for Police Extreme Training Based on TK-Noser Regularization Algorithm[J]..Export: BibTex EndNote
Journal Information
Sponsored by:Institute of Biophysics, The Chinese Academy of Sciences; Biophysical Society of China Edited by: Editorial Office of Progress in Biochemistry and Biophysics Published by:Editorial Office of PIBB Editor-in-Chief:HE Rong-Qiao Adress:15 Datun Road, Chaoyang District,Beijing 100101,China Telephone:86-10-64888459 Email:prog@ibp.ac.cn Journal Inclusion:SCIE, CA, SCOPUS, JST, AJ, Core Journals, CSCD, WJCI ISSN    1000-3282 CN    11-2161/Q Current Issue
External Links
Chinese Academy of SciencesInstitute of Biophysics, Chinese Academy of SciencesBiophysical Society of China