Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (187)

Search Parameters:
Keywords = Gdnf

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 4226 KB  
Review
Molecular Mechanisms of the Microbiota–Gut–Brain Axis in the Onset and Progression of Stroke
by Javier Caballero-Villarraso, Sara Pons-Villarta, Jerónimo Cruces-Párraga, Ainoa Navarrete-Pérez, Antonio Camargo, Juan Antonio Moreno, Isaac Túnez and Eduardo Agüera-Morales
Int. J. Mol. Sci. 2025, 26(20), 10071; https://doi.org/10.3390/ijms262010071 - 16 Oct 2025
Viewed by 454
Abstract
The bidirectional relationship between the brain and gut microbiota has led to the concept of the microbiota–gut–brain axis. It refers to a system of bilateral communication that integrates neuronal, immunological, and metabolic signals, whose disruption has been linked to the pathogenesis of digestive, [...] Read more.
The bidirectional relationship between the brain and gut microbiota has led to the concept of the microbiota–gut–brain axis. It refers to a system of bilateral communication that integrates neuronal, immunological, and metabolic signals, whose disruption has been linked to the pathogenesis of digestive, metabolic, and neurological disorders, among others. Intestinal dysbiosis (an imbalance in the gut microbiota) can promote a proinflammatory and prothrombotic state, as well as dyslipidaemia and dysglycemia, that increase atherogenic risk and consequently the risk of stroke. Dysbiosis can also lead to neuroinflammatory and neurodegenerative effects, compromising the integrity of the blood–brain barrier and exacerbating brain injury after stroke. Specific bacterial profiles have been associated with varying levels of stroke risk, emphasising the role of gut microbiota-derived vasoactive metabolites such as Trimethylamine N-Oxide (TMAO), phenylacetylglnutamine (PAGln), and short-chain fatty acids (SCFAs), which may serve as biomarkers for stroke risk and severity. Gut microbiota also influences neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF), involved in recovery after stroke. Research has explored the potential to modify the gut microbiota to either prevent stroke (by reducing risk) or improve outcomes (by decreasing severity and sequelae). Current scientific evidence supports the role of gut microbiota as a potential diagnostic and prognostic biomarker, as well as a therapeutic target. Full article
Show Figures

Figure 1

13 pages, 982 KB  
Article
Secretome from Uterine Cervical Mesenchymal Stem Cells as a Protector of Neuronal Cells Against Oxidative Stress and Inflammation
by Javier Mateo, Miguel Ángel Suárez-Suárez, Maria Fraile, Ángel Ramón Piñera-Parrilla, Francisco J. Vizoso and Noemi Eiro
Biomolecules 2025, 15(10), 1402; https://doi.org/10.3390/biom15101402 - 2 Oct 2025
Viewed by 387
Abstract
Background: The limited self-repair capacity of nerve tissue requires a new therapeutic approach. Mesenchymal stem cells from the uterine cervix, hUCESC, have shown anti-inflammatory, regenerative, and anti-oxidative stress effects through their secretome, which makes them candidates to evaluate their potential in the context [...] Read more.
Background: The limited self-repair capacity of nerve tissue requires a new therapeutic approach. Mesenchymal stem cells from the uterine cervix, hUCESC, have shown anti-inflammatory, regenerative, and anti-oxidative stress effects through their secretome, which makes them candidates to evaluate their potential in the context of neuronal damage. In this study, we aimed to determine whether secretome or conditioned medium of hUCESC (hUCESC-CM) has beneficial action in the treatment of PC-12 and HMC3 cells in vitro under conditions of oxidative stress and inflammation. Methods: Differentiated PC-12 cells and HMC3 cells were subjected to oxidative stress and inflammatory conditions in the presence of hUCESC-CM. The expression of factors related to both processes was evaluated by q-RT-PCR. Results: PC-12 cells treated with hUCESC-CM showed a significant increase in the expression of anti-oxidative stress factors (HO-1 and Nrf2) and a significant decrease in the expression of pro-inflammatory factors (IL1β, IL6 and TNFα). In addition, the treatment of HMC3 cells with hUCESC-CM significantly decreased the expression of IL6 and TNFα and enhanced the expression of neuroprotective factors such as BDNF and GDNF. Conclusions: Considering that both oxidative stress and inflammation are interrelated and implicated in several nerve injuries and neurodegenerative disorders, the effects of hUCESC-CM on neuronal cells are very promising. Full article
(This article belongs to the Section Biological Factors)
Show Figures

Figure 1

29 pages, 4385 KB  
Review
The Dual Role of Astrocytes in CNS Homeostasis and Dysfunction
by Aarti Tiwari, Satyabrata Rout, Prasanjit Deep, Chandan Sahu and Pradeep Kumar Samal
Neuroglia 2025, 6(4), 38; https://doi.org/10.3390/neuroglia6040038 - 29 Sep 2025
Viewed by 1077
Abstract
Astrocytes are the most common type of glial cell in the central nervous system (CNS). They have many different functions that go beyond just supporting other cells. Astrocytes were once thought of as passive parts of the CNS. However, now they are known [...] Read more.
Astrocytes are the most common type of glial cell in the central nervous system (CNS). They have many different functions that go beyond just supporting other cells. Astrocytes were once thought of as passive parts of the CNS. However, now they are known to be active regulators of homeostasis and active participants in both neurodevelopmental and neurodegenerative processes. This article looks at the both sides of astrocytic function: how they safeguard synaptic integrity, ion and neurotransmitter balance, and blood-brain barrier (BBB) stability, as well as how astrocytes can become activated and participate in the immune response by releasing cytokines, upregulating interferons, and modulating the blood–brain barrier and inflammation disease condition. Astrocytes affect and influence neuronal function through the tripartite synapse, gliotransmission, and the glymphatic system. When someone is suffering from neurological disorders, reactive astrocytes become activated after being triggered by factors such as pro-inflammatory cytokines, chemokines, and inflammatory mediators, these reactive astrocytes, which have higher levels of glial fibrillary acidic protein (GFAP), can cause neuroinflammation, scar formation, and the loss of neurons. This review describes how astrocytes are involved in important CNS illnesses such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis, and ischemia. It also emphasizes how these cells can change from neuroprotective to neurotoxic states depending on the situation. Researchers look at important biochemical pathways, such as those involving toll-like receptors, GLP-1 receptors, and TREM2, to see if they can change how astrocytes respond. Astrocyte-derived substances, including BDNF, GDNF, and IL-10, are also essential for protecting and repairing neurons. Astrocytes interact with other CNS cells, especially microglia and endothelial cells, thereby altering the neuroimmune environment. Learning about the molecular processes that control astrocytic plasticity opens up new ways to treat glial dysfunction. This review focuses on the importance of astrocytes in the normal and abnormal functioning of the CNS, which has a significant impact on the development of neurotherapeutics that focus on glia. Full article
Show Figures

Figure 1

19 pages, 5648 KB  
Article
Role of RET-Regulated GDNF-GFRα1 Endocytosis in Methamphetamine-Induced Neurotoxicity
by Mengran Lv, Baoyu Shen, Zhenling Wu, Genmeng Yang, Yuanyuan Cao, Yuan Zhang, Junjie Shu, Wenjuan Dong, Zhenping Hou, Di Jing, Xinjie Zhang, Yuhan Hou, Jing Xu, Lihua Li and Shijun Hong
Int. J. Mol. Sci. 2025, 26(19), 9522; https://doi.org/10.3390/ijms26199522 - 29 Sep 2025
Viewed by 409
Abstract
Methamphetamine (METH) is a highly addictive synthetic psychostimulant that can induce severe neurotoxicity, leading to neurodegeneration similar to neurodegenerative diseases. The endocytosis of glial cell line-derived neurotrophic factor (GDNF) and its family receptor alpha 1 (GFRα1), regulated by transmembrane receptor tyrosine kinase (RET), [...] Read more.
Methamphetamine (METH) is a highly addictive synthetic psychostimulant that can induce severe neurotoxicity, leading to neurodegeneration similar to neurodegenerative diseases. The endocytosis of glial cell line-derived neurotrophic factor (GDNF) and its family receptor alpha 1 (GFRα1), regulated by transmembrane receptor tyrosine kinase (RET), has been shown to resist neurodegeneration. Specifically, the endocytosis of GDNF-GFRα1 mediated by RET is crucial in protecting neurons. Although many molecular mechanisms of METH induced neurotoxicity have been explored, the obstacles to the neuroprotective effect of GDNF in the context of METH induced neurotoxicity are still unclear. In this study, an increase in cell apoptosis and GDNF expression was observed in the hippocampus of METH abusers. METH also induces cell degeneration, cytotoxicity, and GDNF expression and release in hippocampal neuronal (HT-22) cells in a concentration-dependent manner (0.25, 0.5, 1, 2, and 4 mM) and time-dependent manner (3, 6, 12, 24, and 48 h). Meanwhile, after 24 h of exposure to METH (2mM), apoptosis, impaired endocytosis of GDNF-GFRα1, and decreased expression of RET were observed in HT-22 cells and organotypic hippocampal slices of mice. More notably, overexpression of RET weakened METH induced cell degeneration, apoptosis, and disruption of GDNF-GFRα1 endocytosis in HT-22 cells. This study suggests that RET is a key molecule for METH to disrupt GDNF-mediated neuroprotective signaling, and targeting RET-mediated endocytosis of GDNF-GFRα1 may be a potential therapeutic approach for METH induced neurotoxicity and neurodegeneration. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

30 pages, 1350 KB  
Review
Glial Cells as Emerging Therapeutic Targets in Neurodegenerative Diseases: Mechanistic Insights and Translational Perspectives
by Thirupathirao Vishnumukkala, Che Mohd Nasril Che Mohd Nassir, Zaw Myo Hein, Prarthana Kalerammana Gopalakrishna, Barani Karikalan, Aisyah Alkatiri, Saravanan Jagadeesan, Venkatesh R. Naik, Warren Thomas, Mohamad Aris Mohd Moklas and Mohd Amir Kamaruzzaman
Cells 2025, 14(19), 1497; https://doi.org/10.3390/cells14191497 - 24 Sep 2025
Viewed by 1028
Abstract
Neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease, multiple sclerosis, and amyotrophic lateral sclerosis share converging mechanisms of neuronal dysfunction, including protein aggregation, oxidative stress, and chronic neuroinflammation. Glial cells, once considered passive supporters, are now recognized as central [...] Read more.
Neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease, multiple sclerosis, and amyotrophic lateral sclerosis share converging mechanisms of neuronal dysfunction, including protein aggregation, oxidative stress, and chronic neuroinflammation. Glial cells, once considered passive supporters, are now recognized as central drivers of these processes, offering both pathogenic triggers and therapeutic opportunities. Yet, despite compelling preclinical evidence, the translation of glial-targeted therapies into clinical success has been limited. This review provides a critical synthesis of current knowledge by examining therapeutic strategies through the lens of their translational challenges and failures. This narrative review highlights how interspecies variability of glial phenotypes, shifting neuroprotective versus neurotoxic states, limited biomarker stratification, and delivery barriers have constrained trials, such as anti-triggering receptor expressed on myeloid cells 2 (anti-TREM2) antibodies in AD and glial cell line-derived neurotrophic factor (GDNF) in PD. By analyzing these obstacles across major neurodegenerative disorders, this review argue that the next stage of glial medicine requires precision approaches that integrate stage-specific phenotyping, biomarker-guided patient selection, and innovative delivery platforms. Understanding not only what has been tried but why translation has stalled is essential to chart a roadmap for effective, disease-modifying glial therapies in the aging brain. Full article
(This article belongs to the Special Issue Glial Cells in Aging Neuroscience)
Show Figures

Figure 1

21 pages, 4172 KB  
Review
ASTROGLIA: Molecular Mechanisms, Functional Roles, and Neurophysiological Implications in the Central Nervous System
by Andrea Ortega, Luz A. Martínez-Nuncio, Elisa Taddei, Eduardo Castañeda, Carmen Rubio and Moisés Rubio-Osornio
Life 2025, 15(10), 1505; https://doi.org/10.3390/life15101505 - 24 Sep 2025
Viewed by 438
Abstract
Astrocytes, the principal components of astroglia, play essential roles in maintaining neuronal and synaptic homeostasis in the central nervous system. By regulating extracellular levels of glutamate, potassium (K+), and calcium (Ca2+), they prevent excitotoxicity and support neuronal survival. Astrocytes [...] Read more.
Astrocytes, the principal components of astroglia, play essential roles in maintaining neuronal and synaptic homeostasis in the central nervous system. By regulating extracellular levels of glutamate, potassium (K+), and calcium (Ca2+), they prevent excitotoxicity and support neuronal survival. Astrocytes also modulate synaptic transmission and plasticity through gliotransmission, including vesicular glutamate release and D-serine synthesis via the serine shuttle, which regulates NMDA receptor activity. They provide metabolic support by facilitating glucose and oxygen transport from the vasculature, forming dynamic neurovascular units. Through signaling pathways such as cAMP-PKA and interactions with neurotrophic factors like BDNF and GDNF, astrocytes influence gene expression, synaptic remodeling, and plasticity. Furthermore, astrocytes exhibit regional and functional heterogeneity, which underlies their diverse contributions to both physiological and pathological conditions, including neurodegenerative diseases. This review summarizes current knowledge on astrocytic regulation of neuronal homeostasis, synaptic plasticity, and metabolism, highlighting their mechanisms of network communication, gliotransmission, and regional specialization, and discusses their implications in health and disease. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

17 pages, 1443 KB  
Brief Report
Priming Canine Adipose Tissue-Derived Mesenchymal Stem Cells with CBD-Rich Cannabis Extract Modulates Neurotrophic Factors Expression Profile
by Vinicius Skau Perino, Lucas Vinícius de Oliveira Ferreira, Beatriz da Costa Kamura, Natielly Dias Chimenes, Alisson Vinícius Gimenes Olbera, Thiago Tourinho Pereira, Aline Márcia Marques Braz, Marjorie de Assis Golim, Márcio de Carvalho and Rogério Martins Amorim
Vet. Sci. 2025, 12(10), 926; https://doi.org/10.3390/vetsci12100926 - 24 Sep 2025
Viewed by 479
Abstract
The endocannabinoid system regulates key biological functions such as neuroprotection, pain modulation, inflammation, and immunomodulation. Cannabis-based therapies have gained attention due to the therapeutic potential of their bioactive compounds, particularly phytocannabinoids like cannabidiol (CBD), which exhibit anti-inflammatory, neuroprotective, and immunomodulatory properties. Mesenchymal stem [...] Read more.
The endocannabinoid system regulates key biological functions such as neuroprotection, pain modulation, inflammation, and immunomodulation. Cannabis-based therapies have gained attention due to the therapeutic potential of their bioactive compounds, particularly phytocannabinoids like cannabidiol (CBD), which exhibit anti-inflammatory, neuroprotective, and immunomodulatory properties. Mesenchymal stem cells (MSCs) are widely studied for their regenerative and immunomodulatory potential. This study evaluated the effects of priming canine adipose tissue-derived MSCs (cAT-MSCs) with a CBD-rich cannabis extract on cell morphology, viability, neurotrophic factor gene expression, and cytokine gene and protein expression. cAT-MSCs (n = 5) were primed for 24 h and divided into three groups: Control (C, unprimed), D1 (2.25 µM CBD), and D2 (225 nM CBD). No morphological or viability changes were observed. Gene expression analysis showed that groups D1 and D2 exhibited increased HGF expression. D1 also showed increased IDO and decreased BDNF expression. In contrast, no significant changes were observed in GDNF, IL-10, TNF-α, IFN-γ, or PTGES2. Regarding the cytokine profile, GM-CSF, IL-2, and IL-10 were undetectable. Notably, IL-8 and MCP-1 levels were significantly reduced in D1 compared to the control. These findings suggest that CBD priming modulates key regenerative and inflammatory mediators in cAT-MSCs, supporting its potential application in enhancing the efficacy of cell-based therapies. Full article
(This article belongs to the Special Issue Animal Anesthesia and Analgesia Management: Pain Relief and Sedation)
Show Figures

Figure 1

17 pages, 4780 KB  
Article
The Generation of a Testicular Peritubular Cell Line from Giant Pandas (Ailuropoda melanoleuca)
by Xueni You, Yuren Wang, Yuliang Liu, Rong Hou, Yi Zheng and Junhui An
Cells 2025, 14(18), 1426; https://doi.org/10.3390/cells14181426 - 11 Sep 2025
Viewed by 683
Abstract
Giant pandas (Ailuropoda melanoleuca), a flagship endangered species under priority preservation in China, remain poorly understood in terms of their testicular physiology and the mechanisms underlying spermatogenesis. Testicular peritubular cells (TPTCs), a crucial somatic cell type surrounding seminiferous tubules, secrete growth [...] Read more.
Giant pandas (Ailuropoda melanoleuca), a flagship endangered species under priority preservation in China, remain poorly understood in terms of their testicular physiology and the mechanisms underlying spermatogenesis. Testicular peritubular cells (TPTCs), a crucial somatic cell type surrounding seminiferous tubules, secrete growth factors such as GDNF and CSF1 and release inflammatory factors such as IL-6 and IL-1β, contributing to the testicular niche and immune homeostasis. The contraction of TPTCs also facilitates the transport of sperm towards the epididymis. Nonetheless, TPTCs tend to undergo replicative senescence in vitro, which is a hinderance to their in-depth study. Here, we generated an immortalized monoclonal cell line with TPTC identities from giant pandas via lentiviral transduction of SV40 large T antigen into the cells and the subsequent clonal isolation through limiting dilution. The generated cell line, designated PD-TPTCs, demonstrated unlimited proliferative capacity and has been cultured for over five months and passaged more than 50 times to date. Characterization of PD-TPTCs revealed stable expression of key TPTC markers including ACTA2, MYH11, CNN1, and AR. Moreover, PD-TPTCs could respond to ATP and forskolin (FSK) stimulation with a pro-inflammatory gene expression profile and increased steroidogenic activity, respectively, and they were also amenable to lipofection. As such, the generated PD-TPTC line represents a promising cellular model for future mechanistic studies on the testicular niche, spermatogenesis, and reproductive disorders in giant pandas, laying the foundation for the development of novel assisted reproductive technology (ART) in this endangered species. Full article
(This article belongs to the Special Issue Advances in Reproductive Biology: Cellular and Molecular Mechanisms)
Show Figures

Figure 1

14 pages, 1158 KB  
Article
Neuroinflammatory Signature of Post-Traumatic Confusional State: The Role of Cytokines in Moderate-to-Severe Traumatic Brain Injury
by Federica Piancone, Francesca La Rosa, Ambra Hernis, Ivana Marventano, Pietro Arcuri, Marco Rabuffetti, Jorge Navarro, Marina Saresella, Mario Clerici and Angela Comanducci
Int. J. Mol. Sci. 2025, 26(17), 8593; https://doi.org/10.3390/ijms26178593 - 4 Sep 2025
Viewed by 759
Abstract
Traumatic brain injury (TBI), a leading cause of mortality and disability, recognizes a primary, immediate injury due to external forces, and a secondary phase that includes inflammation that can lead to complications such as the post-traumatic confusional state (PTCS), potentially impacting long-term neurological [...] Read more.
Traumatic brain injury (TBI), a leading cause of mortality and disability, recognizes a primary, immediate injury due to external forces, and a secondary phase that includes inflammation that can lead to complications such as the post-traumatic confusional state (PTCS), potentially impacting long-term neurological recovery. An earlier identification of these complications, including PTCS, upon admission to intensive rehabilitation units (IRU) could possibly allow the design of personalized rehabilitation protocols in the immediate post-acute phase of moderate-to-severe TBI. The present study aims to identify potential biomarkers to distinguish between TBI patients with and without PTCS. We analyzed cellular and molecular mechanisms involved in neuroinflammation (IL-6, IL-1β, IL-10 cytokines), neuroendocrine function (norepinephrine, NE, epinephrine, E, dopamine), and neurogenesis (glial cell line-derived neurotrophic factor, GDNF, insuline-like growth factor 1, IGF-1, nerve growth factor, NGF, brain-derived growth factor, BDNF) using enzyme-linked immunosorbent assay (ELISA), comparing results between 29 TBI patients (17 with PTCS and 12 non-confused) and 34 healthy controls (HC), and correlating results with an actigraphy-derived sleep efficiency parameter. In TBI patients compared to HC, serum concentration of (1) pro-inflammatory IL-1β cytokine was significantly increased while that of anti-inflammatory IL-10 cytokine was significantly decreased; (2) NE, E and DA were significantly increased; (3) GDNF, NGF and IGF-1 were significantly increased while that of BDNF was significantly decreased. Importantly, IL-10 serum concentration was significantly lower in PTCS than in non-confused patients, correlating positively with an improved actigraphy-derived sleep efficiency parameter. An anti-inflammatory environment may be associated with better prognosis after TBI. Full article
Show Figures

Figure 1

24 pages, 1028 KB  
Review
Molecular Links Between Metabolism and Mental Health: Integrative Pathways from GDF15-Mediated Stress Signaling to Brain Energy Homeostasis
by Minju Seo, Seung Yeon Pyeon and Man S. Kim
Int. J. Mol. Sci. 2025, 26(15), 7611; https://doi.org/10.3390/ijms26157611 - 6 Aug 2025
Viewed by 1775
Abstract
The relationship between metabolic dysfunction and mental health disorders is complex and has received increasing attention. This review integrates current research to explore how stress-related growth differentiation factor 15 (GDF15) signaling, ceramides derived from gut microbiota, and mitochondrial dysfunction in the brain interact [...] Read more.
The relationship between metabolic dysfunction and mental health disorders is complex and has received increasing attention. This review integrates current research to explore how stress-related growth differentiation factor 15 (GDF15) signaling, ceramides derived from gut microbiota, and mitochondrial dysfunction in the brain interact to influence both metabolic and psychiatric conditions. Evidence suggests that these pathways converge to regulate brain energy homeostasis through feedback mechanisms involving the autonomic nervous system and the hypothalamic–pituitary–adrenal axis. GDF15 emerges as a key stress-responsive biomarker that links peripheral metabolism with brainstem GDNF family receptor alpha-like (GFRAL)-mediated anxiety circuits. Meanwhile, ceramides impair hippocampal mitochondrial function via membrane incorporation and disruption of the respiratory chain. These disruptions may contribute to sustained pathological states such as depression, anxiety, and cognitive dysfunction. Although direct mechanistic data are limited, integrating these pathways provides a conceptual framework for understanding metabolic–psychiatric comorbidities. Furthermore, differences in age, sex, and genetics may influence these systems, highlighting the need for personalized interventions. Targeting mitochondrial function, GDF15-GFRAL signaling, and gut microbiota composition may offer new therapeutic strategies. This integrative perspective helps conceptualize how metabolic and psychiatric mechanisms interact for understanding the pathophysiology of metabolic and psychiatric comorbidities and highlights therapeutic targets for precision medicine. Full article
Show Figures

Figure 1

18 pages, 3194 KB  
Article
Identification and Characterization of the Complete Genome of the TGF-β Gene Family in Tupaia belangeri: Expression and Function of Adipose Tissue Under Cold Acclimation Conditions
by Lijie Du, Wanlong Zhu and Lin Zhang
Int. J. Mol. Sci. 2025, 26(14), 6681; https://doi.org/10.3390/ijms26146681 - 11 Jul 2025
Viewed by 666
Abstract
The transforming growth factor beta (TGF-β) gene family is widely distributed across the animal kingdom, playing a crucial role in various cellular processes and maintaining overall health and homeostasis. The present study identified 34 TGF-β family genes based on the [...] Read more.
The transforming growth factor beta (TGF-β) gene family is widely distributed across the animal kingdom, playing a crucial role in various cellular processes and maintaining overall health and homeostasis. The present study identified 34 TGF-β family genes based on the genome sequence in Tupaia belangeri, which were classified into the TGF-β, bone morphogenetic protein (BMP), growth differentiation factor (GDF), glial cell-derived neurotrophic factor (GDNF), and Activin/Inhibin subfamilies. A phylogenetic analysis revealed the evolutionary relationships among members of the TGF-β family in T. belangeri and their homologous genes in Homo sapiens, Mus musculus, and Pan troglodytes, indicating a high degree of conservation throughout evolution. A chromosomal distribution and collinearity analysis demonstrated the localization of these genes within the genome of T. belangeri and their collinearity with genes from other species. A gene structure and motif analysis further illustrated the conservation and diversity among TGF-β family members. A protein interaction network analysis highlighted the central roles of TGFB1, TGFB3, BMP7, and BMP2 in signal transduction. A functional enrichment analysis underscored the significance of the TGF-β signaling pathway in the biological processes of T. belangeri, particularly in cell proliferation, differentiation, and apoptosis. We assessed the impact of cold acclimation treatment on the expression of TGF-β family proteins in the adipose tissue (white adipose tissue [WAT] and brown adipose tissue [BAT]) of T. belangeri using ELISA technology, finding that protein expression levels in the experimental group were significantly higher than those of in the control group. These results suggested that cold acclimation may enhance the adaptability of T. belangeri to cold environments by modulating the expression of TGF-β family genes. This study offers new insights into the role of the TGF-β family in the cold acclimation adaptation of T. belangeri, providing a scientific foundation for future genetic improvements and strategies for cold acclimation. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

25 pages, 6990 KB  
Article
Study on the Pharmacological Efficacy and Mechanism of Dual-Target Liposome Complex AD808 Against Alzheimer’s Disease
by Chang Liu, Xiaoqing Wang, Wei Xu, Songli Yu, Yueru Zhang, Qiming Xu and Xiangshi Tan
Pharmaceuticals 2025, 18(7), 977; https://doi.org/10.3390/ph18070977 - 29 Jun 2025
Viewed by 967
Abstract
Background/Objectives: To study the efficacy and pharmacological mechanism of the dual-target liposome complex AD808 in the treatment of Alzheimer’s disease. Methods: Using APP/PS1 mouse models, the therapeutic efficacy and pharmacological mechanism of AD808 on Alzheimer’s disease were studied through water maze [...] Read more.
Background/Objectives: To study the efficacy and pharmacological mechanism of the dual-target liposome complex AD808 in the treatment of Alzheimer’s disease. Methods: Using APP/PS1 mouse models, the therapeutic efficacy and pharmacological mechanism of AD808 on Alzheimer’s disease were studied through water maze tests, brain tissue staining, immunofluorescence, and ELISA for inflammatory and neurotrophic factors. Results: AD808 exhibited significant pharmacodynamic effects in improving behavioral and cognitive abilities (70% reduction in escape latency) and repairing damaged nerve cells (90% reduction in Aβ plaque) in Alzheimer’s disease mice. The efficacy of the liposome complex AD808 was significantly better than that of ST707 or gh625-Zn7MT3 alone. AD808 significantly reduced brain inflammation (57.3% and 61.5% reductions in TNF-α and IL-1β, respectively) in AD (Alzheimer’s disease) mouse models and promoted the upregulation of neurotrophic factors and nerve growth factors (142.8% increase in BDNF, 275.9% in GDNF, and 111.3% in NGF-1) in brain homogenates. By activating the PI3K/AKT signaling pathway in brain microglia, AD808 upregulated TREM2 protein expression and removed Aβ amyloid plaques in the brain. Additionally, it promoted the transition of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, regulated the M1/M2 balance, released anti-inflammatory and neurotrophic factors, reduced chronic inflammation, and enhanced neurological repair. Based on these results, the potential pharmacological mechanism of AD808 against Alzheimer’s disease was proposed. Conclusions: As a dual-target liposome complex, AD808 has shown promising therapeutic potential in the treatment of Alzheimer’s disease, providing a new strategy for innovative drug development. Full article
(This article belongs to the Special Issue Pharmacotherapy for Alzheimer’s Disease)
Show Figures

Figure 1

16 pages, 2167 KB  
Article
Lifestyle and Clinical Predictors of Glial Cell Line-Derived Neurotrophic Factor Expression in Lumbosacral Stenosis-Related Ligamentum Flavum Degeneration
by Dawid Sobański, Małgorzata Sobańska, Rafał Staszkiewicz, Damian Strojny, Werner Dammermann, Paweł Gogol, Weronika Wieczorek-Olcha, Artur Chwalba and Beniamin Oskar Grabarek
Biomedicines 2025, 13(7), 1530; https://doi.org/10.3390/biomedicines13071530 - 23 Jun 2025
Viewed by 3005
Abstract
Background/Objectives: Degenerative spinal conditions, such as degenerative stenosis, have been linked to metabolic and lifestyle factors, including obesity, smoking, and diabetes. Glial cell line-derived neurotrophic factor (GDNF) plays a crucial role in neuroprotection, but its relationship with these risk factors remains unclear. [...] Read more.
Background/Objectives: Degenerative spinal conditions, such as degenerative stenosis, have been linked to metabolic and lifestyle factors, including obesity, smoking, and diabetes. Glial cell line-derived neurotrophic factor (GDNF) plays a crucial role in neuroprotection, but its relationship with these risk factors remains unclear. Methods: This study aims to evaluate the relationship between body mass index (BMI), smoking, diabetes, and GDNF levels in patients with degenerative spine conditions. We measured the GDNF levels in patients with degenerative stenosis and assessed the impact of BMI, smoking status, and the presence of diabetes. Comparisons were made using appropriate statistical analyses to determine the significance of these factors on GDNF levels. Results: A significant inverse relationship was observed between the BMI and GDNF levels (p < 0.01). Patients with a higher BMI exhibited lower GDNF concentrations. Additionally, patients who smoked or had diabetes showed significantly lower GDNF levels compared to non-smokers and those without diabetes (p = 0.03 and p = 0.02, respectively). These findings suggest that both metabolic and lifestyle factors are associated with decreased GDNF, which may accelerate neurodegenerative processes in the spine. Conclusions: Our study demonstrates that increased BMI, smoking, and diabetes are linked to reduced GDNF levels, potentially contributing to the progression of degenerative spine conditions such as stenosis. These findings highlight the need for targeted clinical interventions to manage these risk factors, aiming to preserve GDNF levels and slow the degenerative processes in the spine. Future research should explore therapeutic approaches to modulate GDNF in affected populations. Full article
Show Figures

Figure 1

19 pages, 2936 KB  
Article
Association of Gene Expression Profiles in HPV-Positive Head and Neck Squamous Cell Carcinoma with Patient Outcome: In Search of Prognostic Biomarkers
by J. Noé García-Chávez, Adriana Contreras-Paredes, Claudia González-Espinosa, Imelda Martínez-Ramírez, Elizabeth Langley, Marcela Lizano and J. Omar Muñoz-Bello
Int. J. Mol. Sci. 2025, 26(12), 5894; https://doi.org/10.3390/ijms26125894 - 19 Jun 2025
Viewed by 2164
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) ranks sixth in incidence and seventh in cancer mortality worldwide. Approximately 30% of HNSCC cases are related to human papillomavirus (HPV) infection, the oropharynx being the anatomical subsite most associated with HPV infection. Traditionally, HPV-positive HNSCC [...] Read more.
Head and Neck Squamous Cell Carcinoma (HNSCC) ranks sixth in incidence and seventh in cancer mortality worldwide. Approximately 30% of HNSCC cases are related to human papillomavirus (HPV) infection, the oropharynx being the anatomical subsite most associated with HPV infection. Traditionally, HPV-positive HNSCC has been considered to have better treatment response and clinical outcome. However, HPV-positive HNSCC is a heterogeneous group since 30% of the cases present early relapse, which implies that there are differences in molecular profiles within HPV-positive patients. In this study, we used bioinformatic data analysis from open-access repositories to compare molecular profiles differentially expressed between HPV-positive and -negative HNSCC patients. Using the TCGA HNSCC transcriptomic data, we identified a group of genes, whose expression is related to clinical outcome in patients. Our findings were validated in an independent cohort confirming that the expression levels of FABP4, HMGA2, S100A10, GDNF, SLC7A,2 and GPR18 genes were associated with overall survival (OS) exclusively in HPV-positive HNSCC patients, while ST6GALNAC1 expression was associated with OS in HPV-negative HNSCC. The expression of OS-related genes was independent of tumor stage and history of alcoholism. Our findings suggest that transcriptional profiles in HPV-positive HNSCC are an excellent source of information for the search for potential prognostic biomarkers. Full article
(This article belongs to the Special Issue Viral Infections and Cancer: Recent Advances and Future Perspectives)
Show Figures

Figure 1

15 pages, 3838 KB  
Article
Extended Toxicity, Genotoxicity, and Mutagenicity of Combination of pBudK-coVEGF-coANG and pBudK-coGDNF Plasmids in Preclinical Trials
by Igor V. Samatoshenkov, Alexander M. Aimaletdinov, Elena Y. Zakirova, Egan L. Kalmykov, Rustam Khodzhibaev, Yulia M. Samatoshenkova, Ilnur M. Ganiev, Marat S. Kadyrov and Yana O. Mukhamedshina
Biomedicines 2025, 13(5), 1223; https://doi.org/10.3390/biomedicines13051223 - 18 May 2025
Viewed by 783
Abstract
Chronic lower limb ischemia is a debilitating condition, particularly prevalent among elderly patients and individuals ineligible for revascularization procedures. Gene therapy aimed at promoting therapeutic angiogenesis presents a promising alternative treatment strategy. Objectives: This study evaluated the preclinical safety of a gene therapy [...] Read more.
Chronic lower limb ischemia is a debilitating condition, particularly prevalent among elderly patients and individuals ineligible for revascularization procedures. Gene therapy aimed at promoting therapeutic angiogenesis presents a promising alternative treatment strategy. Objectives: This study evaluated the preclinical safety of a gene therapy drug composed of the plasmids pBudK-coVEGF-coANG and pBudK-coGDNF in laboratory animals. Safety assessment followed a single intramuscular injection at a dose 30 times higher than the proposed therapeutic level. Methods: Acute toxicity was monitored over a 24-h period. Genotoxicity was assessed using the micronucleus test at doses of 200, 1000, and 5000 μg/kg. Bone marrow cytology was analyzed to detect hematopoietic toxicity. Delayed toxicity was evaluated over a two-week recovery period. Results: No signs of acute toxicity were observed, even at the highest dose. The micronucleus test revealed no genotoxic effects, with no significant increase in micronucleated polychromatic erythrocytes compared to control groups. Bone marrow erythroblast parameters remained within normal physiological ranges. Additionally, no delayed adverse effects were detected during the recovery period. Conclusions: The gene therapy drug demonstrated a favorable preclinical safety profile, exhibiting no evidence of toxicity or genotoxicity, even at substantially elevated doses. These findings support the continued development of this therapy as a potential treatment for chronic lower limb ischemia in patients who are not candidates for surgical intervention. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

Back to TopTop