WANG Peng-Xiao , CHEN Le-Rong , WANG Zhen , LONG Jian-Gang , PENG Yun-Hua
2025, 52(7):1649-1660. DOI: 10.16476/j.pibb.20250008 CSTR: 32369.14.pibb.20250008
Abstract:Mitochondria, functioning not only as the central hub of cellular energy metabolism but also as semi-autonomous organelles, orchestrate cellular fate decisions through their endogenous mitochondrial DNA (mtDNA), which encodes core components of the electron transport chain. Emerging research has identified microRNAs localized within mitochondria, termed mitochondria-located microRNAs (mitomiRs). Recent studies have revealed that mitomiRs are transcribed from nuclear DNA (nDNA), processed and matured in the cytoplasm, and subsequently transported into mitochondria. mitomiRs regulate mtDNA through diverse mechanisms, including modulation of mtDNA expression at the translational level and direct binding to mtDNA to influence transcription. Aberrant expression of mitomiRs leads to mitochondrial dysfunction and contributes to the pathogenesis of metabolic diseases. Restoring mitomiR expression to physiological levels using mitomiRs mimics or inhibitors has been shown to improve mitochondrial function and alleviate related diseases. Consequently, the regulatory mechanisms of mitomiRs have become a major focus in mitochondrial research. Given that mitomiRs are located in mitochondria, targeted delivery strategies designed for mtDNA can be adapted for the delivery of mitomiRs mimics or inhibitors. However, numerous intracellular and extracellular barriers remain, highlighting the need for more precise and efficient delivery systems in the future. The regulation of mtDNA expression mediated by mitomiRs not only expands our understanding of miRNA functions in post-transcriptional gene regulation but also provides promising molecular targets for the treatment of mitochondrial-related diseases. This review systematically summarizes recent research progress on mitomiRs in regulating mtDNA expression and discusses the underlying mechanisms of mitomiRs-mtDNA interactions. Additionally, it provides new perspectives on precision therapeutic strategies, with a particular emphasis on mitomiRs-based regulation of mitochondrial function in mitochondrial-related diseases.
CHANG Zhan-Xin , MIAO Long , WANG Peng
2025, 52(7):1661-1672. DOI: 10.16476/j.pibb.20240528 CSTR: 32369.14.pibb.20240528
Abstract:Mitochondria play a pivotal role in spermatogenesis and sperm activation in Caenorhabditis elegans, serving as the primary ATP supplier for cell division and differentiation while also acting as a key regulator of zinc ion homeostasis, membrane dynamics, and apoptotic signaling. This review systematically summarizes the essential mitochondrial mechanisms at different stages of sperm development, highlighting their multifaceted contributions beyond energy metabolism. Mitochondria are crucial for maintaining the health and stability of the gonads by regulating key apoptotic execution proteins that facilitate the proper elimination of damaged or unnecessary germ cells. Additionally, mitochondria dynamically adjust their energy supply to meet the metabolic demands of different stages of germline development. During early spermatogenesis, mitochondria provide ATP to fuel mitotic and meiotic divisions, support cellular differentiation, and regulate H+ and Zn2+ exchange to maintain cytoplasmic homeostasis, thereby ensuring the proper maturation and functionality of sperm cells. As spermatogenesis progresses, mitochondria participate in processing and sorting essential sperm proteins, such as major sperm protein (MSP), and contribute to the formation of membranous organelles (MOs), which are critical for subsequent activation events. During sperm activation, mitochondria play a dual role in ensuring a successful transition from immotile spermatids to fully functional spermatozoa. First, they provide ATP to facilitate pseudopod formation, MO fusion, and ion channel regulation, all of which are essential for sperm motility and fertilization potential. Second, mitochondria regulate the quality and quantity of functional mitochondria within sperm cells through mitopherogenesis—a recently discovered process in which mitochondrial vesicles are selectively released, ensuring that only healthy mitochondria are retained. This quality-control mechanism optimizes mitochondrial function, which is crucial for sustaining sperm motility and longevity. Beyond their traditional role in energy metabolism, mitochondria may also contribute to protein synthesis during spermatogenesis and activation. Recent evidence suggests that mitochondrial ribosomes actively translate specific proteins required for sperm function, challenging the long-standing belief that spermatozoa do not engage in de novo protein synthesis after differentiation. This emerging perspective raises important questions about the role of mitochondria in regulating sperm activation at the molecular level, particularly in modulating oxidative phosphorylation (OXPHOS) protein composition to optimize ATP production. In summary, mitochondria serve as both the central energy hub and a crucial regulatory factor in sperm activation, metabolic homeostasis, and reproductive success. Their involvement extends beyond ATP generation to include apoptotic regulation, ion homeostasis, vesicle-mediated mitochondrial quality control, and potential contributions to protein synthesis. Understanding these mitochondrial functions in C. elegans not only deepens our knowledge of nematode reproductive biology, but also provides valuable insights into broader mechanisms governing mitochondrial regulation in germline cells across species. These findings open new avenues for future research into the interplay between mitochondria, energy metabolism, and sperm function, with potential implications for reproductive health and fertility studies.
FENG Jia-Jia , GUO Meng , OUYANG Zheng , Lü Bin
2025, 52(7):1673-1686. DOI: 10.16476/j.pibb.2024.0451 CSTR: 32369.14.pibb.20240451
Abstract:The liver, skeletal muscle, and adipose tissue are central energy-metabolizing organs and insulin-sensitive tissues, playing a crucial role in maintaining glucose homeostasis. As the powerhouse of the cell, mitochondria not only regulate insulin secretion but also oversee the oxidative phosphorylation and β-oxidation of fatty acids, processes vital for the metabolism of carbohydrates and fats, as well as the synthesis of ATP. The mitochondrial quality control system is of paramount importance for sustaining mitochondrial homeostasis, achieved through mechanisms such as protein homeostasis, mitochondrial dynamics, mitophagy, and biogenesis. Evidence suggests that dysfunctional mitochondria may significantly contribute to insulin resistance and ectopic fat storage in the liver, offering new insights into the strong correlation between mitochondrial dysfunction and the development of obesity, diabetes mellitus type 2 (T2DM), and non-alcoholic fatty liver disease (NAFLD). This manuscript aims to delve into the precise mechanisms by which imbalances in mitochondrial quality control lead to metabolic disorders in the liver, skeletal muscle, and adipose tissue, the 3 major insulin-sensitive organs. In the liver, mitochondrial dysfunction can lead to disturbances in glucose and lipid metabolism, resulting in insulin resistance and fat accumulation—a key factor in the development of NAFLD. In skeletal muscle, reduced mitochondrial function can decrease ATP production, weakening the muscle’s ability to uptake glucose, thereby exacerbating insulin resistance. In adipose tissue, mitochondrial dysfunction can impair adipocyte function, leading to lipotoxicity and inflammatory responses,which further contribute to insulin resistance and the onset of metabolic syndrome. Moreover, the interorgan crosstalk among these 3 tissues is essential for overall metabolic homeostasis. For instance, hepatic gluconeogenesis and glucose utilization in skeletal muscle are both influenced by the health status of their respective mitochondrial populations. The conversion between different types of adipose tissue and the ability to store lipids depend on normal mitochondrial function to avert ectopic fat accumulation in other organs. In summary, this manuscript emphasizes the critical role of mitochondrial quality control in maintaining the metabolic stability of the liver, skeletal muscle, and adipose tissue. It elucidates the specific mechanisms by which mitochondrial dysfunction in these organs contributes to the development of metabolic diseases, providing a foundation for future research and the development of therapeutic strategies targeting mitochondrial dysfunction.
ZHANG Wen-Long , QUAN Lei , ZHAO Yun-Gang
2025, 52(7):1687-1707. DOI: 10.16476/j.pibb.2024.0525 CSTR: 32369.14.pibb.20240525
Abstract:Mitochondria, the primary energy-producing organelles of the cell, also serve as signaling hubs and participate in diverse physiological and pathological processes, including apoptosis, inflammation, oxidative stress, neurodegeneration, and tumorigenesis. As semi-autonomous organelles, mitochondrial functionality relies on nuclear support, with mitochondrial biogenesis and homeostasis being stringently regulated by the nuclear genome. This interdependency forms a bidirectional signaling network that coordinates cellular energy metabolism, gene expression, and functional states. During mitochondrial damage or dysfunction, retrograde signals are transmitted to the nucleus, activating adaptive transcriptional programs that modulate nuclear transcription factors, reshape nuclear gene expression, and reprogram cellular metabolism. This mitochondrion-to-nucleus communication, termed “mitochondrial retrograde signaling”, fundamentally represents a mitochondrial “request” to the nucleus to maintain organellar health, rooted in the semi-autonomous nature of mitochondria. Despite possessing their own genome, the “fragmented” mitochondrial genome necessitates reliance on nuclear regulation. This genomic incompleteness enables mitochondria to sense and respond to cellular and environmental stressors, generating signals that modulate the functions of other organelles, including the nucleus. Evolutionary transfer of mitochondrial genes to the nuclear genome has established mitochondrial control over nuclear activities via retrograde communication. When mitochondrial dysfunction or environmental stress compromises cellular demands, mitochondria issue retrograde signals to solicit nuclear support. Studies demonstrate that mitochondrial retrograde signaling pathways operate in pathological contexts such as oxidative stress, electron transport chain (ETC) impairment, apoptosis, autophagy, vascular tension, and inflammatory responses. Mitochondria-related diseases exhibit marked heterogeneity but invariably result in energy deficits, preferentially affecting high-energy-demand tissues like muscles and the nervous system. Consequently, mitochondrial dysfunction underlies myopathies, neurodegenerative disorders, metabolic diseases, and malignancies. Dysregulated retrograde signaling triggers proliferative and metabolic reprogramming, driving pathological cascades. Mitochondrial retrograde signaling critically influences tumorigenesis and progression. Tumor cells with mitochondrial dysfunction exhibit compensatory upregulation of mitochondrial biogenesis, excessive superoxide production, and ETC overload, collectively promoting metastatic tumor development. Recent studies reveal that mitochondrial retrograde signaling—mediated by altered metabolite levels or stress signals—induces epigenetic modifications and is intricately linked to tumor initiation, malignant progression, and therapeutic resistance. For instance, mitochondrial dysfunction promotes oncogenesis through mechanisms such as epigenetic dysregulation, accumulation of mitochondrial metabolic intermediates, and mitochondrial DNA (mtDNA) release, which activates the cytosolic cGAS-STING signaling pathway. In normal cells, miR-663 mediates mitochondrion-to-nucleus retrograde signaling under reactive oxygen species (ROS) regulation. Mitochondria modulate miR-663 promoter methylation, which governs the expression and supercomplex stability of nuclear-encoded oxidative phosphorylation (OXPHOS) subunits and assembly factors. However, dysfunctional mitochondria induce oxidative stress, elevate methyltransferase activity, and cause miR-663 promoter hypermethylation, suppressing miR-663 expression. Mitochondrial dysfunction also triggers retrograde signaling in primary mitochondrial diseases and contributes to neurodegenerative disorders such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). Current therapeutic strategies targeting mitochondria in neurological diseases focus on 5 main approaches: alleviating oxidative stress, inhibiting mitochondrial fission, enhancing mitochondrial biogenesis, mitochondrial protection, and insulin sensitization. In AD patients, mitochondrial morphological abnormalities and enzymatic defects, such as reduced pyruvate dehydrogenase and α-ketoglutarate dehydrogenase activity, are observed. Platelets and brains of AD patients exhibit diminished cytochrome c oxidase (COX) activity, correlating with mitochondrial dysfunction. To model AD-associated mitochondrial pathology, researchers employ cybrid technology, transferring mtDNA from AD patients into enucleated cells. These cybrids recapitulate AD-related mitochondrial phenotypes, including reduced COX activity, elevated ROS production, oxidative stress markers, disrupted calcium homeostasis, activated stress signaling pathways, diminished mitochondrial membrane potential, apoptotic pathway activation, and increased Aβ42 levels. Furthermore, studies indicate that Aβ aggregates in AD and α-synuclein aggregates in PD trigger mtDNA release from damaged microglial mitochondria, activating the cGAS-STING pathway. This induces a reactive microglial transcriptional state, exacerbating neurodegeneration and cognitive decline. Targeting the cGAS-STING pathway may yield novel therapeutics for neurodegenerative diseases like AD, though translation from bench to bedside remains challenging. Such research not only deepens our understanding of disease mechanisms but also informs future therapeutic strategies. Investigating the triggers, core molecular pathways, and regulatory networks of mitochondrial retrograde signaling advances our comprehension of intracellular communication and unveils novel pathogenic mechanisms underlying malignancies, neurodegenerative diseases, and type 2 diabetes mellitus. This review summarizes established mitochondrial-nuclear retrograde signaling axes, their roles in interorganellar crosstalk, and pathological consequences of dysregulated communication. Targeted modulation of key molecules and proteins within these signaling networks may provide innovative therapeutic avenues for these diseases.
LI Jin-Ru , DUAN Yu , DAI Xin-Gui , YAO Yong-Ming
2025, 52(7):1708-1727. DOI: 10.16476/j.pibb.2024.0487 CSTR: 32369.14.pibb.20240487
Abstract:Interferon stimulating factor STING, a transmembrane protein residing in the endoplasmic reticulum, is extensively involved in the sensing and transduction of intracellular signals and serves as a crucial component of the innate immune system. STING is capable of directly or indirectly responding to abnormal DNA originating from diverse sources within the cytoplasm, thereby fulfilling its classical antiviral and antitumor functions. Structurally, STING is composed of 4 transmembrane helices, a cytoplasmic ligand binding domain (LBD), and a C terminal tail structure (CTT). The transmembrane domain (TM), which is formed by the transmembrane helical structures, anchors STING to the endoplasmic reticulum, while the LBD is in charge of binding to cyclic dinucleotides (CDNs). The classical second messenger, cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), represents a key upstream molecule for STING activation. Once cGAMP binds to LBD, STING experiences conformational alterations, which subsequently lead to the recruitment of Tank-binding kinase 1 (TBK1) via the CTT domain. This, in turn, mediates interferon secretion and promotes the activation and migration of dendritic cells, T cells, and natural killer cells. Additionally, STING is able to activate nuclear factor-κB (NF-κB), thereby initiating the synthesis and release of inflammatory factors and augmenting the body’s immune response. In recent years, an increasing number of studies have disclosed the non-classical functions of STING. It has been found that STING plays a significant role in organelle regulation. STING is not only implicated in the quality control systems of organelles such as mitochondria and endoplasmic reticulum but also modulates the functions of these organelles. For instance, STING can influence key aspects of organelle quality control, including mitochondrial fission and fusion, mitophagy, and endoplasmic reticulum stress. This regulatory effect is not unidirectional; rather, it is subject to organelle feedback regulation, thereby forming a complex interaction network. STING also exerts a monitoring function on the nucleus and ribosomes, which further enhances the role of the cGAS-STING pathway in infection-related immunity. The interaction mechanism between STING and organelles is highly intricate, which, within a certain range, enhances the cells’ capacity to respond to external stimuli and survival pressure. However, once the balance of this interaction is disrupted, it may result in the occurrence and development of inflammatory diseases, such as aseptic inflammation and autoimmune diseases. Excessive activation or malfunction of STING may trigger an over-exuberant inflammatory response, which subsequently leads to tissue damage and pathological states. This review recapitulates the recent interactions between STING and diverse organelles, encompassing its multifarious functions in antiviral, antitumor, organelle regulation, and immune regulation. These investigations not only deepen the comprehension of molecular mechanisms underlying STING but also offer novel concepts for the exploration of human disease pathogenesis and the development of potential treatment strategies. In the future, with further probing into STING function and its regulatory mechanisms, it is anticipated to pioneer new approaches for the treatment of complex diseases such as inflammatory diseases and tumors.
WANG Qing-Qing , LIU Ya , LIU Wei , LONG Wei
2025, 52(7):1728-1744. DOI: 10.16476/j.pibb.2024.0509 CSTR: 32369.14.pibb.20240509
Abstract:Radiation-induced injury is a key factor in determining the prognosis of patients undergoing radiotherapy, highlighting the significant clinical importance of developing drugs for radiation prevention and treatment. Especially in oncology, radiation-induced injury remains a pivotal determinant of therapeutic outcomes, because of its direct correlation with normal tissue damage during radiotherapy. Efforts to mitigate or treat such injury are thus paramount in enhancing the overall safety and efficacy of cancer treatment. Novel nanomedicines with prolonged systemic circulation, versatile drug-loading capacities, enhanced tissue retention, and stimuli responsiveness exhibit unique advantages in the treatment and prevention of radiation-induced diseases, as they can be designed based on the specific microenvironment of radiation-damaged tissues, which offers innovative solutions to address the limitations of conventional radioprotectors such as short half-life, poor tissue targeting, and systemic side effects. This review thus aims to provide an overview of recent advance in the design and application of nanomaterials for radiation prevention and treatment. Generally, ionizing radiation damages cells either by inducing DNA double-strand breaks or through the generation of reactive oxygen species (ROS). The resulting oxidative stress would disrupt the structural integrity of cell membranes, proteins, and nucleic acids, leading to apoptosis, chronic inflammation, and systemic effects across multiple systems, including hematopoietic system, gastrointestinal tract, skin, lungs, brain, and heart. Radiation protection strategies focus on scavenging ROS, stimulating cellular repair and regeneration, inducing tissue hypoxia, and inhibiting apoptotic pathways. Recent advances in nanomedicine have introduced novel approaches for targeted and efficient radiation protection and treatment. For radiation-induced hematopoietic injury, nanoparticles can been designed to promote red and white blood cell regeneration while reducing oxidative stress. To address radiation-induced gastrointestinal injuries, nanomaterials enable localized antioxidant delivery and extended intestinal retention, effectively relieving radiation enteritis by scavenging ROS and modulating gut microbiota. For radiation-induced skin injuries, self-assembling peptide hydrogels that mimic the extracellular matrix can serve as effective scaffolds for wound healing. These hydrogels exhibit excellent antioxidant properties, stimulating angiogenesis, and accelerating the recovery of radiation dermatitis. In cases of radiation-induced brain damage, nanoparticles were designed to cross the blood-brain barrier to rescue neuronal damage and protect cognitive function. This review provides an in-depth insight into the mechanisms underlying radiation-induced injuries and highlights how nanomaterial were construtced according to the specific injury. Therefore, nanotechnology endowers durgs with transformative potential for preventing and treating radiation-induced injuries. Despite significant progress in nanomedicine, there are still challenges in long-term biocompatibility, precise targeting of damaged tissues, and scalable manufacturing. In addition, an in-depth understanding of the interactions between nanomaterials and biological systems remains to be covered. Future efforts should focus on optimizing design strategies, enhancing clinical translatability, and ensuring long-term safety, ultimately improving patient outcomes. Besides, expanding research into other radiation-induced diseases, such as radiation-induced ophthalmic disorders and hepatic injuries, may diversify therapeutic options.
SHI Wan-Rui , CUI Li-Gang , LIANG Xiao-Long
2025, 52(7):1745-1756. DOI: 10.16476/j.pibb.2025.0037 CSTR: 32369.14.pibb.20250037
Abstract:Pancreatic cancers (PCs) is a common malignant tumor with poor prognosis in the digestive system. Its main treatment methods include surgery, radiotherapy, chemotherapy, and targeted therapy. The early diagnosis rate of hidden onset of PCs is low, and most patients have already lost the opportunity to undergo surgery when diagnosed with PCs. Chemotherapy is still the main treatment for advanced PCs, but the use of chemotherapy drugs in PCs can easily lead to drug resistance. The most significant feature that distinguishes PCs from other tumors is its rich and dense matrix, which not only hinders drug penetration but also impedes the infiltration of immune cells. The above reasons have led to a very low survival rate of PCs patients. Therefore, drug delivery systems are very important in the diagnosis and treatment of PCs. They can improve drug delivery, enhance biological barrier penetration, reduce side effects, and combine multiple treatment methods. Therefore, the treatment prospects of PCs are very broad. Currently, drug delivery systems widely applied in PCs primarily include nanodrug delivery systems, tumor microenvironment-targeted drug delivery system, immunotherapy drug delivery system, gene therapy drug delivery system, and combination therapy drug delivery system that synergize multiple therapeutic modalities. Emerging drug delivery systems (DDSs) have revolutionized PCs treatment by addressing these challenges through multiple mechanisms. Nanoformulations improve drug solubility, prolong circulation time, and reduce systemic toxicity via passive/active targeting. Smart DDSs responsive to PCs-specific stimuli enable extracellular matrix degradation, tumor-associated fibroblasts reprogramming, and vascular normalization to enhance drug accessibility. Last but not least, carrier systems loaded with myeloid-derived suppressor cell inhibitors or T cell activators can reverse immunosuppression and potentiate immunotherapy efficacy. Advanced platforms co-deliver chemotherapeutics with immunomodulators, gene-editing tools, or sonodynamic agents to achieve synergistic antitumor effects. These platforms aim to address critical challenges in PCs treatment, such as enhancing drug bioavailability, overcoming stromal barriers, reprogramming immunosuppressive niches, and achieving multi-mechanistic antitumor effects. This article provides a systematic summary and prospective analysis of the current development status, latest cutting-edge advances, opportunities, and challenges of the above-mentioned drug delivery systems in the field of PCs therapy.
CHEN Meng-Meng , HU Nan , BAO Shuang-Qing , LI Xiao-Hong
2025, 52(7):1757-1770. DOI: 10.16476/j.pibb.2024.0488 CSTR: 32369.14.pibb.20240488
Abstract:Brain organoids are three-dimensional (3D) neural cultures that self-organize from pluripotent stem cells (PSCs) cultured in vitro. Compared with traditional two-dimensional (2D) neural cell culture systems, brain organoids demonstrate a significantly enhanced capacity to faithfully replicate key aspects of the human brain, including cellular diversity, 3D tissue architecture, and functional neural network activity. Importantly, they also overcome the inherent limitations of animal models, which often differ from human biology in terms of genetic background and brain structure. Owing to these advantages, brain organoids have emerged as a powerful tool for recapitulating human-specific developmental processes, disease mechanisms, and pharmacological responses, thereby providing an indispensable model for advancing our understanding of human brain development and neurological disorders. Despite their considerable potential, conventional brain organoids face a critical limitation: the absence of a functional vascular system. This deficiency results in inadequate oxygen and nutrient delivery to the core regions of the organoid, ultimately constraining long-term viability and functional maturation. Moreover, the lack of early neurovascular interactions prevents these models from fully recapitulating the human brain microenvironment. In recent years, the introduction of vascularization strategies has significantly enhanced the physiological relevance of brain organoid models. Researchers have successfully developed various vascularized brain organoid models through multiple innovative approaches. Biological methods, for example, involve co-culturing brain organoids with endothelial cells to induce the formation of static vascular networks. Alternatively, co-differentiation strategies direct both mesodermal and ectodermal lineages to generate vascularized tissues, while fusion techniques combine pre-formed vascular organoids with brain organoids. Beyond biological approaches, tissue engineering techniques have played a pivotal role in promoting vascularization. Microfluidic systems enable the creation of dynamic, perfusable vascular networks that mimic blood flow, while 3D printing technologies allow for the precise fabrication of artificial vascular scaffolds tailored to the organoid’s architecture. Additionally, in vivo transplantation strategies facilitate the formation of functional, blood-perfused vascular networks through host-derived vascular infiltration. The incorporation of vascularization has yielded multiple benefits for brain organoid models. It alleviates hypoxia within the organoid core, thereby improving cell survival and supporting long-term culture and maturation. Furthermore, vascularized organoids recapitulate critical features of the neurovascular unit, including the early structural and functional characteristics of the blood-brain barrier. These advancements have established vascularized brain organoids as a highly relevant platform for studying neurovascular disorders, drug screening, and other applications. However, achieving sustained, long-term functional perfusion while preserving vascular structural integrity and promoting vascular maturation remains a major challenge in the field. In this review, we systematically outline the key stages of human neurovascular development and provide a comprehensive analysis of the various strategies employed to construct vascularized brain organoids. We further present a detailed comparative assessment of different vascularization techniques, highlighting their respective strengths and limitations. Additionally, we summarize the principal challenges currently faced in brain organoid vascularization and discuss the specific technical obstacles that persist. Finally, in the outlook section, we elaborate on the promising applications of vascularized brain organoids in disease modeling and drug testing, address the main controversies and unresolved questions in the field, and propose potential directions for future research.
TANG Xiao-Xia , ZHANG Shu-Jia , ZHANG Ying , WANG Li
2025, 52(7):1771-1791. DOI: 10.16476/j.pibb.20250024 CSTR: 32369.14.pibb.20250024
Abstract:Self-face is a unique and highly distinctive stimulus, not shared with others, and serves as a reliable marker of self-awareness. Compared to other faces, self-face processing exhibits several advantages, including the self-face recognition advantage, self-face attention advantage, and self-face positive processing advantage. The self-face recognition advantage manifests as faster and more accurate identification across different orientations and spatial frequency components, supported by enhanced early event-related potential (ERP) components, such as N170. Attentional biases toward self-face are evident in target detection during spatial tasks and the attentional blink effect in temporal paradigms. However, measurement sensitivity, perceptual load, and task demands contribute to some mixed findings. Positive biases further characterize the self-face processing advantage, with individuals perceiving their faces as more attractive or trustworthy than objective representations. These biases even extend to self-similar others, influencing social behaviors such as trust and voting preferences. Self-face processing advantages have been observed at an unconscious level and are regulated by several factors, including self-esteem, cultural differences, and multisensory integration. Cultural and individual differences play a crucial role in shaping self-face advantages. Individuals from Western cultures, which emphasize independent self-construal, exhibit stronger self-face biases compared to those from East Asian collectivist contexts. Self-esteem also modulates self-face advantages: high-self-esteem individuals generally maintain their self-face recognition advantage despite interference, exhibit attentional prioritization of self-faces, and demonstrate enhanced positive associations with subliminal self-faces. In contrast, low-self-esteem individuals display recognition vulnerabilities to social cues, show context-dependent attentional divergence (prioritizing others’ faces in task-oriented settings while prioritizing self-face in free-viewing tasks), and exhibit reversed positive associations with subliminal self-faces. Multisensory integration, such as synchronized visual-tactile cues, enhances self-face advantages and induces perceptual plasticity. This phenomenon is exemplified by the enfacement illusion, in which synchronous visual and tactile inputs update the mental representation of the self-face, leading to assimilation with another face. Neuroanatomically, self-face processing is predominantly lateralized to the right hemisphere and involves a network of brain regions, including the occipital lobe, temporal lobe, frontal lobe, insula, and cingulate gyrus. Disruptions in these networks are linked to self-face processing deficits in socio-cognitive disorders. For instance, autism spectrum disorder (ASD) and schizophrenia are associated with attenuated self-face advantages and abnormal neural activity in regions such as the right inferior frontal gyrus, insula, and posterior cingulate cortex. These findings suggest that self-face processing could serve as a potential biomarker for the early diagnosis and intervention of such disorders. In recent years, researchers have proposed various theoretical explanations for self-face processing and its advantage effects. However, some studies have reported no significant behavioral or neural advantages of self-faces over familiar faces, leaving the specificity of self-face a subject of debate. Further elucidation of self-face specificity requires the adoption of a face association paradigm, which controls for facial familiarity and helps determine whether qualitative differences exist between self-faces and familiar faces. Given the close relationship between self-face processing advantages and socio-cognitive disorders (e.g., ASD, schizophrenia), a deeper understanding of self-face specificity has the potential to provide critical insights into the early identification, classification, and intervention of these disorders. This research holds both theoretical significance and substantial social value.
XING Wen-Xiao , LUO Fu-Cheng , Lü Tao
2025, 52(7):1792-1803. DOI: 10.16476/j.pibb.2024.0496 CSTR: 32369.14.pibb.20240496
Abstract:In the central nervous system (CNS), the myelin sheath, a specialized membrane structure that wraps around axons, is formed by oligodendrocytes through a highly coordinated spatiotemporal developmental program. The process begins with the directed differentiation of neural precursor cells into oligodendrocyte precursor cells (OPCs), followed by their migration, proliferation, differentiation, and maturation, ultimately leading to the formation of a multi-segmental myelin sheath structure. Recent single-cell sequencing research has revealed that this process involves the temporal regulation of over 200 key genes, with a regulatory network composed of transcription factors such as Sox10 and Olig2 playing a central role. The primary function of the myelin sheath is to accelerate nerve signal transmission and protect nerve fibers from damage. Its insulating properties not only increase nerve conduction speed by 50-100 times but also ensure the long-term functional integrity of the nervous system by maintaining axonal metabolic homeostasis and providing mechanical protection. The pathological effects of myelin sheath injury exhibit a cascade amplification pattern: acute demyelination leads to action potential conduction block, while chronic lesions may cause axonal damage and neuronal death in severe or long-term cases, ultimately resulting in irreversible neurological dysfunction with neurodegenerative characteristics. Multiple sclerosis (MS) is a neurodegenerative disease characterized by chronic inflammatory demyelination of the CNS. Clinically, the distribution of lesions in MS exhibits spatial heterogeneity, which is closely related to differences in the regenerative capacity of oligodendrocytes within the local microenvironment. Emerging evidence suggests that astrocytes form a dynamic “neural-immune-metabolic interface” and play a multidimensional regulatory role in myelin development and regeneration by forming heterogeneous populations composed of different subtypes. During embryonic development, astrocytes induce the targeted differentiation of OPCs in the ventricular region through the Wnt/β-catenin pathway. In the mature stage, they secrete platelet-derived growth factor AA (PDGF-AA) to establish a chemical gradient that guides the precise migration of OPCs along axonal bundles. Notably, astrocytes also provide crucial metabolic support by supplying energy substrates for high-energy myelin formation through the lactate shuttle mechanism. In addition, astrocytes play a dual role in myelin regulation. During the acute injury phase, reactive astrocytes establish a triple defense system within 72 h: upregulating glial fibrillary acidic protein (GFAP) to form scars that isolate lesions, activating the JAK-STAT3 regeneration pathway in oligodendrocytes via leukemia inhibitory factor (LIF), and releasing tumor necrosis factor-stimulated gene-6 (TSG-6) to inhibit excessive microglial activation. However, in chronic neurodegenerative diseases, the phenotypic transformation of astrocytes contributes to microenvironmental deterioration. The secretion of chondroitin sulfate proteoglycans (CSPGs) inhibits OPC migration via the RhoA/ROCK pathway, while the persistent release of reactive oxygen species (ROS) leads to mitochondrial dysfunction and the upregulation of complement C3-mediated synaptic pruning. This article reviews the mechanisms by which astrocytes regulate the development and regeneration of myelin sheaths in the CNS, with a focus on analyzing the multifaceted roles of astrocytes in this process. It emphasizes that astrocytes serve as central hubs in maintaining myelin homeostasis by establishing a metabolic microenvironment and signaling network, aiming to provide new therapeutic strategies for neurodegenerative diseases such as multiple sclerosis.
GUO Xing-Chen , XIE Yan , WEI Xin-Shuo , LI Wen-Fen , SUN Ying-Yu
2025, 52(7):1804-1816. DOI: 10.16476/j.pibb.2024.0393 CSTR: 32369.14.pibb.20240393
Abstract:Locomotion, a fundamental motor function encompassing various forms such as swimming, walking, running, and flying, is essential for animal survival and adaptation. The mesencephalic locomotor region (MLR), located at the midbrain-hindbrain junction, is a conserved brain area critical for controlling locomotion. This review highlights recent advances in understanding the MLR’s structure and function across species, from lampreys to mammals and birds, with a particular focus on insights gained from optogenetic studies in mammals. The goal is to uncover universal strategies for MLR-mediated locomotor control. Electrical stimulation of the MLR in species such as lampreys, salamanders, cats, and mice initiates locomotion and modulates speed and patterns. For example, in lampreys, MLR stimulation induces swimming, with increased intensity or frequency enhancing propulsive force. Similarly, in salamanders, graded stimulation transitions locomotor outputs from walking to swimming. Histochemical studies reveal that effective MLR stimulation sites colocalize with cholinergic neurons, suggesting a conserved neurochemical basis for locomotion control. In mammals, the MLR comprises two key nuclei: the cuneiform nucleus (CnF) and the pedunculopontine nucleus (PPN). Both nuclei contain glutamatergic and GABAergic neurons, with the PPN additionally housing cholinergic neurons. Optogenetic studies in mice by selectively activating glutamatergic neurons have demonstrated that the CnF and PPN play distinct roles in motor control: the CnF drives rapid escape behaviors, while the PPN regulates slower, exploratory movements. This functional specialization within the MLR allows animals to adapt their locomotion patterns and speed in response to environmental demands and behavioral objectives. Similar to findings in lampreys, the CnF and PPN in mice transmit motor commands to spinal effector circuits by modulating the activity of brainstem reticular formation neurons. However, they achieve this through distinct reticulospinal pathways, enabling the generation of specific behaviors. Further insights from monosynaptic rabies viral tracing reveal that the CnF and PPN integrate inputs from diverse brain regions to produce context-appropriate behaviors. For instance, glutamatergic neurons in the PPN receive signals from other midbrain structures, the basal ganglia, and medullary nuclei, whereas glutamatergic neurons in the CnF rarely receive inputs from the basal ganglia but instead are strongly influenced by the periaqueductal grey and inferior colliculus within the midbrain. These differential connectivity patterns underscore the specialized roles of the CnF and PPN in motor control, highlighting their unique contributions to coordinating locomotion. Birds exhibit exceptional flight capabilities, yet the avian MLR remains poorly understood. Comparative studies suggest that the pedunculopontine tegmental nucleus (PPTg) in birds is homologous to the mammalian PPN, which contains cholinergic neurons, while the intercollicular nucleus (ICo) or nucleus isthmi pars magnocellularis (ImC) may correspond to the CnF. These findings provide important clues for identifying the avian MLR and elucidating its role in flight control. However, functional validation through targeted experiments is urgently needed to confirm these hypotheses. Optogenetics and other advanced techniques in mice have greatly advanced MLR research, enabling precise manipulation of specific neuronal populations. Future studies should extend these methods to other species, particularly birds, to explore unique locomotor adaptations. Comparative analyses of MLR structure and function across species will deepen our understanding of the conserved and evolved features of motor control, revealing fundamental principles of locomotion regulation throughout evolution. By integrating findings from diverse species, we can uncover how the MLR has been adapted to meet the locomotor demands of different environments, from aquatic to aerial habitats.
SHEN Ruo-Bing , SHEN Wen-Wen , GAO Shu-Gui
2025, 52(7):1817-1832. DOI: 10.16476/j.pibb.20240292 CSTR: 32369.14.pibb.20240292
Abstract:Modified electro-convulsive therapy (MECT) is one of the most potent treatments for major depressive disorder (MDD). However, it remains a second-line option due to significant side effects, such as transient memory loss. The relationship between therapeutic efficacy and cognitive impairment warrants further investigation to develop improved treatment regimens. In this review, we examine recent evidence from magnetic resonance imaging (MRI) studies aiming to identify structural and functional brain changes specifically associated with both the antidepressant effects and the amnesic outcomes of MECT. MECT induces widespread alterations across multiple brain systems. Increases in gray matter volume (GMV) have been observed in the prefrontal, temporal, and parietal cortices, as well as in subcortical regions such as the hippocampus (HP), amygdala, and striatum. Strengthening of myelination has also been reported along the dorsolateral prefrontal-limbic pathways. Functional changes include increased spontaneous neural activity in prefrontal areas, reorganization of intrinsic connectivity within the default mode network (DMN), and altered functional connectivity (FC) among the DMN, salience network (SN), and central executive network (CEN). Correlational studies have identified structural and functional alterations linked to antidepressant efficacy, including right hippocampal volume enlargement, prefrontal cortical thickening, reduced iron deposition in the striatum, decreased FC within certain DMN nodes, and enhanced effective connectivity from the dorsolateral prefrontal cortex (DLPFC) to the right angular gyrus. In contrast, the amnesic effects have been associated with increased volumes in the left hippocampus and bilateral dentate gyrus; enhanced FC in the left angular gyrus and left posterior cingulate cortex (PCC); increased FC between the right ventral anterior insula and DLPFC; and reduced FC in the left thalamus and bilateral precuneus. Changes in the hippocampus appear to correlate with both antidepressant efficacy and memory impairment. Clinical studies have found no significant correlation between the severity of memory impairment and the reduction in depressive symptoms, suggesting that the therapeutic and adverse effects may arise from distinct regional or subregional mechanisms. Supporting this hypothesis, recent findings show that increased right hippocampal volume is significantly associated with reduced depression scores, whereas increased volume in the left dentate gyrus correlates with declines in delayed recall performance. Additionally, enhanced connectivity between the anterior hippocampus and middle occipital gyrus (MOG) has been linked to mood improvement, while decreased FC between the mid-hippocampus and angular gyrus has been associated with impairments in memory integration. In conclusion, current evidence suggests that the antidepressant and memory-impairing effects of MECT may localize to distinct hippocampal subregions. These effects likely result from differential modulation of local neural activity and functional connectivity, leading to divergent behavioral outcomes. Given that both effects may originate in deep and spatially constrained structures such as the hippocampus, small-sample studies and conventional methodologies may fail to differentiate them effectively. Future research should employ large-scale, longitudinal designs utilizing high-field MRI and multimodal neuroimaging to characterize MECT-induced structure-function coupling in the hippocampus and its integration at the network level. Additionally, multiscale analyses spanning molecular, circuit, and network dimensions would be beneficial.
HE Dan , LI Shi-Guo , ZHAN Ai-Bin
2025, 52(7):1833-1852. DOI: 10.16476/j.pibb.2025.0040 CSTR: 32369.14.pibb.20250040
Abstract:Aquatic organisms can secrete biomacromolecules through specialized organs, tissues, or structures, enabling adhesion to underwater material surfaces and leading to severe biofouling issues. This phenomenon adversely impacts aquatic ecosystem health and human activities. Biofouling has emerged as an emerging global environmental challenge. Adhesion serves as the foundation of biofouling, representing a critical step toward a comprehensive understanding of the adhesion mechanisms of aquatic organisms. Biomacromolecules, including proteins, lipids, and carbohydrates, are the primary functional components in the adhesive substances of aquatic fouling organisms. Research indicates that these biomacromolecules exhibit diversity in types and characteristics across different aquatic organisms, yet their adhesion mechanisms show unifying features. Despite significant progress, there remains a lack of comprehensive reviews on the adhesion mechanisms mediated by biomacromolecules in aquatic fouling organisms, particularly on the roles of lipids and carbohydrates. Through a comprehensive analysis of existing literature, this review systematically summarizes the mechanistic roles of three classes of macromolecules in aquatic biofouling adhesion processes. Proteins demonstrate central functionality in interfacial adhesion and cohesion through specialized functional amino acids, conserved structural domains, and post-translational modifications. Lipids enhance structural stability via hydrophobic barrier formation and antioxidative protection mechanisms. Carbohydrates contribute to adhesion persistence through cohesive reinforcement and enzymatic resistance of adhesive matrices. Building upon these mechanisms, this review proposes four prospective research directions: optimization of protein-mediated adhesion functionality, elucidation of lipid participation in adhesion dynamics, systematic characterization of carbohydrate adhesion modalities, and investigation of macromolecular synergy in composite adhesive systems. The synthesized knowledge provides critical insights into underwater adhesion mechanisms of aquatic fouling organisms and establishes a theoretical foundation for developing mechanism-driven antifouling strategies. This work advances fundamental understanding of bioadhesion phenomena while offering practical guidance for next-generation antifouling technology development.
ZHANG Chang-Jian , LI Yu-Fang , WU Feng-Yun , JIN Rui , NIU Chang , YE Qi-Nong , CHENG Long
2025, 52(7):1853-1865. DOI: 10.16476/j.pibb.2025.0005 CSTR: 32369.14.pibb.20250005
Abstract:Objective The nucleolar protein PES1 (Pescadillo homolog 1) plays critical roles in ribosome biogenesis and cell cycle regulation, yet its involvement in cellular senescence remains poorly understood. This study aimed to comprehensively investigate the functional consequences of PES1 suppression in cellular senescence and elucidate the molecular mechanisms underlying its regulatory role.Methods Initially, we assessed PES1 expression patterns in two distinct senescence models: replicative senescent mouse embryonic fibroblasts (MEFs) and doxorubicin-induced senescent human hepatocellular carcinoma HepG2 cells. Subsequently, PES1 expression was specifically downregulated using siRNA-mediated knockdown in these cell lines as well as additional relevant cell types. Cellular proliferation and senescence were assessed by EdU incorporation and SA-β-gal staining assays, respectively. The expression of senescence-associated proteins (p53, p21, and Rb) and SASP factors (IL-6, IL-1β, and IL-8) were analyzed by Western blot or qPCR. Furthermore, Northern blot and immunofluorescence were employed to evaluate pre-rRNA processing and nucleolar morphology.Results PES1 expression was significantly downregulated in senescent MEFs and HepG2 cells. PES1 knockdown resulted in decreased EdU-positive cells and increased SA-β-gal-positive cells, indicating proliferation inhibition and senescence induction. Mechanistically, PES1 suppression activated the p53-p21 pathway without affecting Rb expression, while upregulating IL-6, IL-1β, and IL-8 production. Notably, PES1 depletion impaired pre-rRNA maturation and induced nucleolar stress, as evidenced by aberrant nucleolar morphology.Conclusion Our findings demonstrate that PES1 deficiency triggers nucleolar stress and promotes p53-dependent (but Rb-independent) cellular senescence, highlighting its crucial role in maintaining nucleolar homeostasis and regulating senescence-associated pathways.
LI Fang , CHEN Bai , WU Yang , LIU Kai , ZHOU Tong , YAO Jia-Feng
2025, 52(7):1866-1877. DOI: 10.16476/j.pibb.20240452 CSTR: 32369.14.pibb.20240452
Abstract:Objective This paper proposes a novel real-time bedside pulmonary ventilation monitoring method for the diagnosis of chronic obstructive pulmonary disease (COPD), based on electrical impedance tomography (EIT). Four indicators—center of ventilation (CoV), global inhomogeneity index (GI), regional ventilation delay inhomogeneity (RVDI), and the ratio of forced expiratory volume in one second to forced vital capacity (FEV1/FVC)—are calculated to enable the spatiotemporal assessment of COPD.Methods A simulation of the respiratory cycles of COPD patients was first conducted, revealing significant differences in certain indicators compared to healthy individuals. The effectiveness of these indicators was then validated through experiments. A total of 93 subjects underwent multiple pulmonary function tests (PFTs) alongside simultaneous EIT measurements. Ventilation heterogeneity under different breathing patterns—including forced exhalation, forced inhalation, and quiet tidal breathing—was compared. EIT images and related indicators were analyzed to distinguish healthy individuals across different age groups from COPD patients.Results Simulation results demonstrated significant differences in CoV, GI, FEV1/FVC, and RVDI between COPD patients and healthy individuals. Experimental findings indicated that, in terms of spatial heterogeneity, the GI values of COPD patients were significantly higher than those of the other two groups, while no significant differences were observed among healthy individuals. Regarding temporal heterogeneity, COPD patients exhibited significantly higher RVDI values than the other groups during both quiet breathing and forced inhalation. Moreover, during forced exhalation, the distribution of FEV1/FVC values further highlighted the temporal delay heterogeneity of regional lung function in COPD patients, distinguishing them from healthy individuals of various ages.Conclusion EIT technology effectively reveals the spatiotemporal heterogeneity of regional lung function, which holds great promise for the diagnosis and management of COPD.
ZHANG Ming-Chen , ZHANG Hui , LI Ting-Ting , CHEN Ming-Hua , WANG Xiao-Wen , SUN Zhong-Guang
2025, 52(7):1878-1889. DOI: 10.16476/j.pibb.2024.0239 CSTR: 32369.14.pibb.20240239
Abstract:Objective The aim of this study was to investigate the prophylactic effects of caloric restriction (CR) on lipopolysaccharide (LPS)-induced septic cardiomyopathy (SCM) and to elucidate the mechanisms underlying the cardioprotective actions of CR. This research aims to provide innovative strategies and theoretical support for the prevention of SCM.Methods A total of forty-eight 8-week-old male C57BL/6 mice, weighing between 20-25 g, were randomly assigned to 4 distinct groups, each consisting of 12 mice. The groups were designated as follows: CON (control), LPS, CR, and CR+LPS. Prior to the initiation of the CR protocol, the CR and CR+LPS groups underwent a 2-week acclimatization period during which individual food consumption was measured. The initial week of CR intervention was set at 80% of the baseline intake, followed by a reduction to 60% for the subsequent 5 weeks. After 6-week CR intervention, all 4 groups received an intraperitoneal injection of either normal saline or LPS (10 mg/kg). Twelve hours post-injection, heart function was assessed, and subsequently, heart and blood samples were collected. Serum inflammatory markers were quantified using enzyme-linked immunosorbent assay (ELISA). The serum myocardial enzyme spectrum was analyzed using an automated biochemical instrument. Myocardial tissue sections underwent hematoxylin and eosin (HE) staining and immunofluorescence (IF) staining. Western blot analysis was used to detect the expression of protein in myocardial tissue, including inflammatory markers (TNF-α, IL-9, IL-18), oxidative stress markers (iNOS, SOD2), pro-apoptotic markers (Bax/Bcl-2 ratio, CASP3), and SIRT3/SIRT6.Results Twelve hours after LPS injection, there was a significant decrease in ejection fraction (EF) and fractional shortening (FS) ratios, along with a notable increase in left ventricular end-systolic diameter (LVESD). Morphological and serum indicators (AST, LDH, CK, and CK-MB) indicated that LPS injection could induce myocardial structural disorders and myocardial injury. Furthermore, 6-week CR effectively prevented the myocardial injury. LPS injection also significantly increased the circulating inflammatory levels (IL-1β, TNF-α) in mice. IF and Western blot analyses revealed that LPS injection significantly up-regulating the expression of inflammatory-related proteins (TNF-α, IL-9, IL-18), oxidative stress-related proteins (iNOS, SOD2) and apoptotic proteins (Bax/Bcl-2 ratio, CASP3) in myocardial tissue. 6-week CR intervention significantly reduced circulating inflammatory levels and downregulated the expression of inflammatory, oxidative stress-related proteins and pro-apoptotic level in myocardial tissue. Additionally, LPS injection significantly downregulated the expression of SIRT3 and SIRT6 proteins in myocardial tissue, and CR intervention could restore the expression of SIRT3 proteins.Conclusion A 6-week CR could prevent LPS-induced septic cardiomyopathy, including cardiac function decline, myocardial structural damage, inflammation, oxidative stress, and apoptosis. The mechanism may be associated with the regulation of SIRT3 expression in myocardial tissue.
FU Ling-Di , DOU Jia-Xuan , YING Ting-Ting , YIN Li-Yong , TANG Min , LIANG Zhen-Hu
2025, 52(7):1890-1903. DOI: 10.16476/j.pibb.2024.0521 CSTR: 32369.14.pibb.20240521
Abstract:Objective Functional near-infrared spectroscopy (fNIRS), a novel non-invasive technique for monitoring cerebral activity, can be integrated with upper limb rehabilitation robots to facilitate the real-time assessment of neurological rehabilitation outcomes. The rehabilitation robot is designed with 3 training modes: passive, active, and resistance. Among these, the resistance mode has been demonstrated to yield superior rehabilitative outcomes for patients with a certain level of muscle strength. The control modes in the resistance mode can be categorized into dynamic and static control. However, the effects of different control modes in the resistance mode on the motor function of patients with upper limb hemiplegia in stroke remain unclear. Furthermore, the effects of force, an important parameter of different control modes, on the activation of brain regions have rarely been reported. This study investigates the effects of dynamic and static resistance modes under varying resistance levels on cerebral functional alterations during motor rehabilitation in post-stroke patients.Methods A cohort of 20 stroke patients with upper limb dysfunction was enrolled in the study, completing preparatory adaptive training followed by 3 intensity-level tasks across 2 motor paradigms. The bilateral prefrontal cortices (PFC), bilateral primary motor cortices (M1), bilateral primary somatosensory cortices (S1), and bilateral premotor and supplementary motor cortices (PM) were examined in both the resting and motor training states. The lateralization index (LI), phase locking value (PLV), network metrics were employed to examine cortical activation patterns and topological properties of brain connectivity.Results The data indicated that both dynamic and static modes resulted in significantly greater activation of the contralateral M1 area and the ipsilateral PM area when compared to the resting state. The static patterns demonstrated a more pronounced activation in the contralateral M1 in comparison to the dynamic patterns. The results of brain network analysis revealed significant differences between the dynamic and resting states in the contralateral PFC area and contralateral M1 area (F=4.709, P=0.038), as well as in the contralateral PM area and ipsilateral M1 area (F=4.218, P=0.049). Moreover, the findings indicated a positive correlation between the activation of the M1 region and the increase in force in the dynamic mode, which was reversed in the static mode.Conclusion Both dynamic and static resistance training modes have been demonstrated to activate the corresponding brain functional regions. Dynamic resistance modes elicit greater oxygen changes and connectivity to the region of interest (ROI) than static resistance modes. Furthermore, the effects of increasing force differ between the two modes. In patients who have suffered a stroke, dynamic modes may have a more pronounced effect on the activation of exercise-related functional brain regions.
MA Hao-Yun , WEI Yu-Yin , HU Li-Ping
2025, 52(7):1904-1908. DOI: 10.16476/j.pibb.2025.0077 CSTR: 32369.14.pibb.20250077
Abstract:The subcortical visual pathway is generally thought to be involved in dangerous information processing, such as fear processing and defensive behavior. A recent study, published in Human Brain Mapping, shows a new function of the subcortical pathway involved in the fast processing of non-emotional object perception. Rapid object processing is a critical function of visual system. Topological perception theory proposes that the initial perception of objects begins with the extraction of topological property (TP). However, the mechanism of rapid TP processing remains unclear. The researchers investigated the subcortical mechanism of TP processing with transcranial magnetic stimulation (TMS). They find that a subcortical magnocellular pathway is responsible for the early processing of TP, and this subcortical processing of TP accelerates object recognition. Based on their findings, we propose a novel training approach called subcortical magnocellular pathway training (SMPT), aimed at improving the efficiency of the subcortical M pathway to restore visual and attentional functions in disorders associated with subcortical pathway dysfunction.
Scan code to follow ® 2025 Website Copyright ICP:京ICP备05023138号-1 京公网安备 11010502031771号
