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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (451)

Search Parameters:
Keywords = nerve cell regeneration

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2443 KiB  
Article
Contralateral Structure and Molecular Response to Severe Unilateral Brain Injury
by Xixian Liao, Xiaojian Xu, Ming Li, Runfa Tian, Yuan Zhuang and Guoyi Gao
Brain Sci. 2025, 15(8), 837; https://doi.org/10.3390/brainsci15080837 (registering DOI) - 5 Aug 2025
Abstract
Background: Severe damage to one side of the brain often leads to adverse consequences and can also cause widespread changes throughout the brain, especially in the contralateral area. Studying molecular changes in the contralateral cerebral hemisphere, especially with regard to genetic regulation, [...] Read more.
Background: Severe damage to one side of the brain often leads to adverse consequences and can also cause widespread changes throughout the brain, especially in the contralateral area. Studying molecular changes in the contralateral cerebral hemisphere, especially with regard to genetic regulation, can help discover potential treatment strategies to promote recovery after severe brain trauma on one side. Methods: In our study, the right motor cortex was surgically removed to simulate severe unilateral brain injury, and changes in glial cells and synaptic structure in the contralateral cortex were subsequently assessed through immunohistological, morphological, and Western blot analyses. We conducted transcriptomic studies to explore changes in gene expression levels associated with the inflammatory response. Results: Seven days after corticotomy, levels of reactive astrocytes and hypertrophic microglia increased significantly in the experimental group, while synapsin-1 and PSD-95 levels in the contralateral motor cortex increased. These molecular changes are associated with structural changes, including destruction of dendritic structures and the encapsulation of astrocytes by synapses. Genome-wide transcriptome analysis showed a significant increase in gene pathways involved in inflammatory responses, synaptic activity, and nerve fiber regeneration in the contralateral cortex after corticorectomy. Key transcription factors such as NF-κB1, Rela, STAT3 and Jun were identified as potential regulators of these contralateral changes. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) confirmed that the mRNA expression levels of Cacna1c, Tgfb1 and Slc2a1 genes related to STAT3, JUN, and NF-κB regulation significantly increased in the contralateral cortex of the experimental group. Conclusions: After unilateral brain damage occurs, changes in the contralateral cerebral hemisphere are closely related to processes involving inflammation and synaptic function. Full article
Show Figures

Figure 1

19 pages, 2574 KiB  
Article
The Neuroregenerative Effects of IncobotulinumtoxinA (Inco/A) in a Nerve Lesion Model of the Rat
by Oscar Sánchez-Carranza, Wojciech Danysz, Klaus Fink, Maarten Ruitenberg, Andreas Gravius and Jens Nagel
Int. J. Mol. Sci. 2025, 26(15), 7482; https://doi.org/10.3390/ijms26157482 - 2 Aug 2025
Viewed by 235
Abstract
The use of Botulinum Neurotoxin A (BoNT/A) to treat peripheral neuropathic pain from nerve injury has garnered interest for its long-lasting effects and safety. This study examined the effects of IncobotulinumtoxinA (Inco/A), a BoNT/A variant without accessory proteins, on nerve regeneration in rats [...] Read more.
The use of Botulinum Neurotoxin A (BoNT/A) to treat peripheral neuropathic pain from nerve injury has garnered interest for its long-lasting effects and safety. This study examined the effects of IncobotulinumtoxinA (Inco/A), a BoNT/A variant without accessory proteins, on nerve regeneration in rats using the chronic constriction injury (CCI) model. Inco/A was administered perineurally at two time points: on days 0 and 21 post CCI. Functional and histological assessments were conducted to evaluate the effect of Inco/A on nerve regeneration. Sciatic Functional Index (SFI) measurements and Compound Muscle Action Potential (CMAP) recordings were conducted at different time points following CCI. Inco/A-treated animals exhibited a 65% improved SFI and 22% reduction in CMAP onset latencies compared to the vehicle-treated group, suggesting accelerated functional nerve recovery. Tissue analysis revealed enhanced remyelination in Inco/A-treated animals and 60% reduction in CGRP and double S100β signal expression compared to controls. Strikingly, 30% reduced immune cell influx into the injury site was observed following Inco/A treatment, suggesting that its anti-inflammatory effect contributes to nerve regeneration. These findings show that two injections of Inco/A promote functional recovery by enhancing neuroregeneration and modulating inflammatory processes, supporting the hypothesis that Inco/A has a neuroprotective and restorative role in nerve injury conditions. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

15 pages, 4965 KiB  
Article
The Rapid Activation of MYDGF Is Critical for Cell Survival in the Acute Phase of Retinal Regeneration in Fish
by Kayo Sugitani, Yuya Omori, Takumi Mokuya, Serika Hosoi, Haruto Kobayashi, Koki Miyata, Yuhei Araiso and Yoshiki Koriyama
Int. J. Mol. Sci. 2025, 26(15), 7251; https://doi.org/10.3390/ijms26157251 - 27 Jul 2025
Viewed by 213
Abstract
Myeloid-derived growth factor (MYDGF), named in reference to its secretion from myeloid cells in bone marrow, is a novel protein with anti-apoptotic and tissue-repairing properties. MYDGF is found in various human tissues affected by different diseases. To date, however, MYDGF expression has yet [...] Read more.
Myeloid-derived growth factor (MYDGF), named in reference to its secretion from myeloid cells in bone marrow, is a novel protein with anti-apoptotic and tissue-repairing properties. MYDGF is found in various human tissues affected by different diseases. To date, however, MYDGF expression has yet to be reported in the nervous system. Herein, we demonstrate for the first time that MYDGF mRNA levels increased in the zebrafish retina 1 h after optic nerve injury (ONI). MYDGF-producing cells were located in the photoreceptors and infiltrating leukocytic cells. We prepared the retina for MYDGF gene knockdown by performing intraocular injections using either MYDGF-specific morpholino or the CRISPR/Cas9 system. Under these MYDGF-knockdown retinal conditions, anti-apoptotic Bcl-2 mRNA was suppressed; in comparison, apoptotic caspase-3 and inflammatory TNFα mRNA were significantly upregulated in the zebrafish retina after ONI compared to the control. Furthermore, heat shock factor 1 (HSF1) was evidently suppressed under these conditions, leading to a significant number of apoptotic neurons. These findings indicate that MYDGF is a key molecule in the stimulation of neuronal regeneration in the central nervous system. Full article
Show Figures

Figure 1

20 pages, 5747 KiB  
Article
Functional Study of the BMP Signaling Pathway in Appendage Regeneration of Exopalaemon carinicauda
by Chaofan Xing, Yong Li, Zhenxiang Chen, Qingyuan Hu, Jiayi Sun, Huanyu Chen, Qi Zou, Yingying Li, Fei Yu, Chao Wang, Panpan Wang and Xin Shen
Biology 2025, 14(8), 940; https://doi.org/10.3390/biology14080940 - 25 Jul 2025
Viewed by 407
Abstract
Appendage autotomy frequently occurs during the cultivation of Exopalaemon carinicauda, which severely impacts its survival and economic benefits. To investigate the molecular mechanism underlying appendage regeneration in E. carinicauda, this study presents a comparative transcriptome analysis on samples from different stages [...] Read more.
Appendage autotomy frequently occurs during the cultivation of Exopalaemon carinicauda, which severely impacts its survival and economic benefits. To investigate the molecular mechanism underlying appendage regeneration in E. carinicauda, this study presents a comparative transcriptome analysis on samples from different stages of appendage regeneration in individuals of the same family of E. carinicauda. A total of 6460 differentially expressed genes (DEGs) were identified between the samples collected at 0 h post-autotomy (D0) and those collected at 18 h post-autotomy (D18h). Additionally, 7740 DEGs were identified between D0 and 14 d post-autotomy (D14d), with 3382 DEGs identified between D18h and D14d. Among them, differentially expressed genes such as EcR, RXR, BMP1, and Smad4 are related to muscle growth or molting and may be involved in the regeneration process. qRT-PCR results revealed that EcBMPR2 was expressed at relatively high levels in the gonad and ventral nerve cord tissues and that the highest level of expression was detected in the regenerative basal tissue at 24 h post-autotomy. In situ hybridization results indicated strong signals of this gene in the cells at the wound site at 72 h post-autotomy. Following knockdown of EcBMPR2, the expression levels of both EcBMPR1B and EcSmad1 were significantly downregulated, and long-term interference with the EcBMPR2 gene resulted in a significantly slower appendage regeneration process compared to the control group. When the downstream transcription factor EcSmad1 was knocked down, the two receptor genes EcBMPR2 and EcBMPR1B were downregulated, whereas EcBMP7 was upregulated. After inhibiting the BMP signaling pathway, the degree of cell aggregation at the autotomy site in the experimental group was significantly lower than that in the control group, the wound healing rate was delayed, and the blastema regeneration time was prolonged from 5 d to 7 d. Collectively, these results indicate that the BMP signaling pathway plays a critical role in the early stages of appendage regeneration in E. carinicauda. This study provides important theoretical insights for understanding limb regeneration in crustaceans. Full article
(This article belongs to the Section Physiology)
Show Figures

Figure 1

19 pages, 3181 KiB  
Article
Overexpression of BDNF and uPA Combined with the Suppression of Von Hippel–Lindau Tumor Suppressor Enhances the Neuroprotective Activity of the Secretome of Human Mesenchymal Stromal Cells in the Model of Intracerebral Hemorrhage
by Stalik S. Dzhauari, Alexandra L. Primak, Nataliya A. Basalova, Natalia I. Kalinina, Anna O. Monakova, Kirill D. Bozov, Arkadiy Ya. Velichko, Maria E. Illarionova, Olga A. Grigorieva, Zhanna A. Akopyan, Vladimir S. Popov, Pavel G. Malkov, Anastasia Yu. Efimenko, Vsevolod A. Tkachuk and Maxim N. Karagyaur
Int. J. Mol. Sci. 2025, 26(14), 6697; https://doi.org/10.3390/ijms26146697 - 12 Jul 2025
Viewed by 377
Abstract
Nerve tissue damage is an unsolved problem in modern neurology and neurosurgery, which prompts the need to search for approaches to stimulate neuroprotection and regeneration of neural tissue. Earlier we have shown that the secretome of human mesenchymal stromal cells (MSCs) stimulates rat [...] Read more.
Nerve tissue damage is an unsolved problem in modern neurology and neurosurgery, which prompts the need to search for approaches to stimulate neuroprotection and regeneration of neural tissue. Earlier we have shown that the secretome of human mesenchymal stromal cells (MSCs) stimulates rat survival, reduces the severity of neurological deficits, and decreases the volume of brain damage in a hemorrhagic stroke model. A significant disadvantage of using the MSC secretome is the need to concentrate it (at least 5–10 fold) to achieve appreciable pharmacological activity. This increases the cost of obtaining clinically applicable amounts of secretome and slows down the clinical translation of this technology. Here, we created a number of genetically modified human MSC cultures, including immortalized MSCs and those with hyperexpression of brain-derived neurotrophic factor (BDNF) and urokinase-type plasminogen activator (uPA) and with suppressed expression of Von Hippel–Lindau tumor suppressor (VHL), and we evaluated the pharmacological activity of their secretomes in a model of intracerebral hemorrhage (ICH) in rats. The secretome of MSCs immortalized by hyperexpression of the catalytic subunit of human telomerase (hTERT) revealed neuroprotective activity indistinguishable from that of primary MSC cultures, yet it still required 10-fold concentration to achieve neuroprotective efficacy. The secretome of MSC culture with combined hyperexpression of BDNF and uPA and suppressed expression of Von Hippel–Lindau tumor suppressor even without additional concentration reduced the severity of neurological disorders and decreased brain lesion volume in the ICH model. The secretomes of MSCs with separate overexpression of BDNF and uPA or suppression of VHL had no such effect or, on the contrary, revealed a toxic effect in the ICH model. Presumably, this may be due to an imbalance in the representation of individual growth factors in the secretome of genetically modified MSCs, which individually may lead to undesirable effects in damaged nervous tissue, such as increased permeability of the blood–brain barrier (under the influence of pro-angiogenic factors) or neural cell apoptosis (due to an excess of neurotrophic factors). The obtained data show that genetic modification of MSC cultures can enhance or alter the therapeutic activity of their secretomes, which can be used in the creation of promising sources of biopharmaceutical substances. Full article
Show Figures

Figure 1

36 pages, 2739 KiB  
Review
Advanced Bioactive Polymers and Materials for Nerve Repair: Strategies and Mechanistic Insights
by Nidhi Puranik, Shraddha Tiwari, Meenakshi Kumari, Shiv Kumar Yadav, Thakur Dhakal and Minseok Song
J. Funct. Biomater. 2025, 16(7), 255; https://doi.org/10.3390/jfb16070255 - 9 Jul 2025
Viewed by 1076
Abstract
Bioactive materials have recently shown potential in nerve repair and regeneration by promoting the growth of new cells, tissue repair, and restoring nerve function. These natural, synthetic, and hybrid materials offer a biomimetic structure, enhance cell attachment, and release bioactive molecules that promote [...] Read more.
Bioactive materials have recently shown potential in nerve repair and regeneration by promoting the growth of new cells, tissue repair, and restoring nerve function. These natural, synthetic, and hybrid materials offer a biomimetic structure, enhance cell attachment, and release bioactive molecules that promote the axonal extension of severed nerves. Scaffold-based preclinical studies have shown promising results on enhancing nerve repair; however, they are limited by the immune response and fabrication, scalability, and cost. Nevertheless, advances in manufacturing, including 3D bioprinting, and other strategies, such as gene editing by CRISPR, will overcome these shortcomings. The opportunity for the development of individualized approaches and specific treatment plans for each patient will also increase the effectiveness of bioactive materials for the treatment of nerve injuries. Combining bioactive materials with the neural interface can develop new reliable therapeutic solutions, particularly for neuroprosthetics. Finally, it is essential to stress a multidisciplinary focus, and future studies are needed to enhance the potential of bioactive materials for patients with nerve injuries and the field of regenerative medicine. Full article
(This article belongs to the Special Issue Active Biomedical Materials and Their Applications, 2nd Edition)
Show Figures

Figure 1

17 pages, 13222 KiB  
Article
Limited Myelination Capacity in Human Schwann Cells in Experimental Models in Comparison to Rodent and Porcine Schwann Cells
by Tak-Ho Chu and Rajiv Midha
Int. J. Mol. Sci. 2025, 26(13), 6457; https://doi.org/10.3390/ijms26136457 - 4 Jul 2025
Viewed by 378
Abstract
Schwann cells (SCs) play a crucial role in peripheral nerve repair by supporting axonal regeneration and remyelination. While extensive research has been conducted using rodent SCs, increasing attention is being directed toward human SCs due to species-specific differences in phenotypical and functional properties, [...] Read more.
Schwann cells (SCs) play a crucial role in peripheral nerve repair by supporting axonal regeneration and remyelination. While extensive research has been conducted using rodent SCs, increasing attention is being directed toward human SCs due to species-specific differences in phenotypical and functional properties, and accessibility of human SCs derived from diverse sources. A major challenge in translating SC-based therapies for nerve repair lies in the inability to replicate human SC myelination in vitro, posing a significant obstacle to drug discovery and preclinical research. In this study, we compared the myelination capacity of human, rodent, and porcine SCs in various co-culture conditions, including species-matched and cross-species neuronal environments in a serum-free medium. Our results confirmed that rodent and porcine SCs readily myelinate neurites under standard culture conditions after treatment with ascorbic acid for two weeks, whereas human SCs, at least within the four-week observation period, failed to show myelin staining in all co-cultures. Furthermore, we investigated whether cell culture manipulation impairs human SC myelination by transplanting freshly harvested and predegenerated human nerve segments into NOD-SCID mice for four weeks. Despite supporting host axonal regeneration into the grafts, human SCs exhibited very limited myelination, suggesting an intrinsic species-specific restriction rather than a cell culture-induced defect. These observations suggest fundamental differences between human and rodent SCs and highlight the need for human-specific models and protocols to advance our understanding of SC myelination. Full article
(This article belongs to the Special Issue Plasticity of the Nervous System after Injury: 2nd Edition)
Show Figures

Figure 1

22 pages, 4242 KiB  
Review
Extracellular Vesicle Metabolomics Holds Promise for Adult Axon Regeneration
by Maria D. Cabrera Gonzalez, Jackson Watson, Laura Leal, Isabella Moceri, Camille Plummer, Biraj Mahato, Abdelrahman Y. Fouda and Sanjoy K. Bhattacharya
Metabolites 2025, 15(7), 454; https://doi.org/10.3390/metabo15070454 - 4 Jul 2025
Viewed by 777
Abstract
Extracellular vesicles (EVs) are bilayer lipid membrane particles that are released by every cell type. These secretions are further classified as exosomes, ectosomes, and microvesicles. They contain biomolecules (RNAs, proteins, metabolites, and lipids) with the ability to modulate various biological processes and have [...] Read more.
Extracellular vesicles (EVs) are bilayer lipid membrane particles that are released by every cell type. These secretions are further classified as exosomes, ectosomes, and microvesicles. They contain biomolecules (RNAs, proteins, metabolites, and lipids) with the ability to modulate various biological processes and have been shown to play a role in intercellular communication and cellular rejuvenation. Various studies suggest exosomes and/or microvesicles as a potential platform for drug delivery. EVs may deliver lipids and nucleotides directly to an injury site in an axon, promoting growth cone stabilization and membrane expansion as well as repair, thus positively modulating adult axon regeneration. In this review, we will provide a perspective on the metabolite composition of EVs in adult axonal regeneration relevant to the central nervous system (CNS), specifically that pertaining to the optic nerve. We will present an overview of the methods for isolation, enrichment, omics data analysis and quantification of extracellular vesicles with the goal of providing direction for future studies relevant to axon regeneration. We will also include current resources for multi-omics data integration relevant to extracellular vesicles from diverse cell types. Full article
Show Figures

Graphical abstract

13 pages, 3604 KiB  
Article
β2-Microglobulin Regulates Extracellular Matrix Dynamics During Peripheral Nerve Injury
by Eiki Shirasawa, Kentaro Uchida, Kenji Onuma, Gen Inoue, Koji Eshima, Masashi Satoh, Masayuki Miyagi, Yoji Toyomura, Akira Norisugi and Masashi Takaso
NeuroSci 2025, 6(3), 59; https://doi.org/10.3390/neurosci6030059 - 29 Jun 2025
Viewed by 479
Abstract
Peripheral nerve injury initiates a complex cascade of events coordinating immune responses, extracellular matrix (ECM) remodeling, and neuronal repair. While β2-microglobulin (B2M) is well known for its role in MHC class I-mediated antigen presentation and CD8+ T-cell differentiation, its potential contributions to [...] Read more.
Peripheral nerve injury initiates a complex cascade of events coordinating immune responses, extracellular matrix (ECM) remodeling, and neuronal repair. While β2-microglobulin (B2M) is well known for its role in MHC class I-mediated antigen presentation and CD8+ T-cell differentiation, its potential contributions to non-immune processes remain underexplored. In this study, we investigated the role of B2M in peripheral nerve regeneration using a chronic constriction injury (CCI) model in wild-type and B2M-deficient (B2M-KO) mice. Flow cytometry, RNA sequencing (RNA-seq), and quantitative PCR (qPCR) were performed to assess T-cell subset dynamics and gene expression following injury. Flow cytometric analysis showed that CD3+CD4+ and CD3+CD8+ T-cell populations increased by day 7 post-injury. While CD3+CD4+ T-cell expansion occurred in both groups, a significant increase in CD3+CD8+ T cells was observed only in wild-type mice. RNA-seq analysis at 3 days post-injury—prior to substantial T-cell accumulation—revealed marked downregulation of ECM-related genes in B2M-KO mice, including collagens, matrix-associated proteins, and other key ECM components. KEGG analysis identified suppression of ECM–receptor interaction, PI3K-Akt, and TGF-β signaling pathways. qPCR confirmed reduced expression of Thbs1 in B2M-KO mice. These findings suggest that B2M plays a critical, CD8+ T-cell-independent role in regulating ECM dynamics and regenerative signaling during early nerve repair, expanding the conceptual framework of B2M’s function beyond classical immune roles. Full article
Show Figures

Figure 1

18 pages, 3351 KiB  
Article
Biosynthesis of Bioactive Human Neurotrophic Factor 3 in Silkworms and Its Biomedical Applications
by Wenjing Geng, Liang Lu, Tangmin Li, Mingyi Zhou, Wei Chen, Hao Tan, Debin Zhong, Guanwang Shen, Ping Lin, Qingyou Xia, Ping Zhao and Zhiqing Li
Insects 2025, 16(7), 676; https://doi.org/10.3390/insects16070676 - 27 Jun 2025
Viewed by 455
Abstract
Neurotrophic factor 3 (NT-3), a potent neurotrophin, promotes neuronal survival and axonal regeneration while demonstrating a unique capacity to induce lineage-specific differentiation of pluripotent stem cells into functional neurons, underscoring its therapeutic potential in neural repair. Despite these advantages, the large-scale production of [...] Read more.
Neurotrophic factor 3 (NT-3), a potent neurotrophin, promotes neuronal survival and axonal regeneration while demonstrating a unique capacity to induce lineage-specific differentiation of pluripotent stem cells into functional neurons, underscoring its therapeutic potential in neural repair. Despite these advantages, the large-scale production of recombinant human NT-3 with preserved structure integrity and functional bioactivity remains a critical challenge. This study takes advantage of the silk gland bioreactor of silkworms for the recombinant expression of human NT-3 protein on a large scale. Our findings reveal that NT-3 was successfully expressed in the middle silk gland of silkworms and secreted into the silk fibers, achieving a yield of up to 0.5 mg of bioactive NT-3 per gram of cocoon weight. The engineered NT-3-functionalized silk material demonstrates no cytotoxicity and significantly enhanced the proliferation, migration, and differentiation of neural cells compared to natural silk protein. Importantly, this functionalized material also promotes neurite outgrowth in HT-22 cells. These results collectively underscore the high bioactivity of the recombinant human NT-3 protein produced in the silkworm silk gland. The ongoing fabrication of NT-3-incorporated silk-based materials holds considerable promise for advancing tissue engineering and nerve regeneration applications. Full article
(This article belongs to the Special Issue Recent Studies on Resource Insects)
Show Figures

Figure 1

21 pages, 1236 KiB  
Review
Gelatin-Based Hydrogels for Peripheral Nerve Regeneration: A Multifunctional Vehicle for Cellular, Molecular, and Pharmacological Therapy
by Denisa-Madalina Viezuina, Irina Musa, Madalina Aldea, Irina-Mihaela Matache, Alexandra-Daniela Rotaru Zavaleanu, Andrei Gresita, Sfredel Veronica and Smaranda Ioana Mitran
Gels 2025, 11(7), 490; https://doi.org/10.3390/gels11070490 - 25 Jun 2025
Viewed by 1017
Abstract
Peripheral nerve injuries (PNIs) present a significant clinical challenge due to the inherently limited regenerative capacity of the adult nervous system. Conventional therapeutic strategies, such as nerve autografting and systemic pharmacological interventions, are often limited by donor site morbidity, restricted graft availability, and [...] Read more.
Peripheral nerve injuries (PNIs) present a significant clinical challenge due to the inherently limited regenerative capacity of the adult nervous system. Conventional therapeutic strategies, such as nerve autografting and systemic pharmacological interventions, are often limited by donor site morbidity, restricted graft availability, and suboptimal drug bioavailability. In this context, gelatin-based hydrogels have emerged as a promising class of biomaterials due to their excellent biocompatibility, biodegradability, and structural similarity to the native extracellular matrix. These hydrogels could offer a highly tunable platform capable of supporting cellular adhesion, promoting axonal elongation, and enabling localized and sustained release of therapeutic agents. This narrative review synthesizes recent advances in the application of gelatin-based hydrogels for peripheral nerve regeneration, with a particular focus on their use as delivery vehicles for neurotrophic factors, stem cells, and pharmacologically active compounds. Additionally, this review provides a foundation for extending our ongoing preclinical study, evaluating the neuroregenerative effects of alpha-lipoic acid, B-complex vitamins, and a deproteinized hemoderivative in a murine PNI model. Although systemic administration has demonstrated promising neuroprotective effects, limitations related to local drug availability and off-target exposure highlight the need for site-specific delivery strategies. In this regard, gelatin hydrogels might represent an excellent candidate for localized, controlled drug delivery. The review concludes by discussing formulation techniques, manufacturing considerations, biological performance, and key translational and regulatory aspects. Full article
(This article belongs to the Special Issue Gelatin-Based Materials for Tissue Engineering)
Show Figures

Figure 1

24 pages, 3669 KiB  
Review
Advances in Polypyrrole Nanofiber Composites: Design, Synthesis, and Performance in Tissue Engineering
by Lu Hao, Demei Yu, Xinyu Hou and Yixuan Zhao
Materials 2025, 18(13), 2965; https://doi.org/10.3390/ma18132965 - 23 Jun 2025
Viewed by 499
Abstract
This review is different from previous studies focusing on polypyrrole (PPy) in universal fields such as sensors and supercapacitors. It is the first TO systematically review the specific applications of PPy-based electrospun nanofiber composites in the biomedical field, focusing on its biocompatibility regulation [...] Read more.
This review is different from previous studies focusing on polypyrrole (PPy) in universal fields such as sensors and supercapacitors. It is the first TO systematically review the specific applications of PPy-based electrospun nanofiber composites in the biomedical field, focusing on its biocompatibility regulation mechanism and tissue repair function. Although PPy exhibits exceptional electrical conductivity, redox activity, and biocompatibility, its clinical translation is hindered by processing challenges and poor degradability. These limitations can be significantly mitigated through composite strategies with degradable nanomaterials, enhancing both process compatibility and biofunctionality. Leveraging the morphological similarity between electrospun nanofibers and the natural extracellular matrix (ECM), this work comprehensively analyzes the topological characteristics of three composite fiber architectures—randomly distributed, aligned, and core–shell structures—and elucidates their application mechanisms in nerve regeneration, skin repair, bone mineralization, and myocardial tissue reconstruction (e.g., facilitating oriented cell migration and regulating differentiation through specific signaling pathway activation). The study further highlights critical challenges in the field, including PPy’s poor solubility, limited spinnability, insufficient mechanical strength, and scalability limitations. Future efforts should prioritize the development of multifunctional gradient composites, intelligent dynamic-responsive scaffolds, and standardized biosafety evaluation systems to accelerate the substantive translation of these materials into clinical applications. Full article
Show Figures

Figure 1

15 pages, 4096 KiB  
Article
Fs-Laser-Induced Micro- and Nanostructures on Polycarbonate and Cellulose Acetate Butyrate for Cell Alignment
by Lukas Wagner, Werner Baumgartner, Agnes Weth, Sebastian Lifka and Johannes Heitz
Appl. Sci. 2025, 15(12), 6754; https://doi.org/10.3390/app15126754 - 16 Jun 2025
Viewed by 389
Abstract
Laser-generated structures have a huge potential to induce an alignment of biological cells, which may be important for various fields in medicine and biotechnology. We describe the formation of fs-laser-induced micro- and nanostructures for achieving the directed growth of Schwann cells, a type [...] Read more.
Laser-generated structures have a huge potential to induce an alignment of biological cells, which may be important for various fields in medicine and biotechnology. We describe the formation of fs-laser-induced micro- and nanostructures for achieving the directed growth of Schwann cells, a type of glial cell that can support the regeneration of nerve pathways by guiding the neuronal axons in the direction of their alignment. Polymer surfaces, i.e., polycarbonate (PC) or cellulose acetate butyrate (CAB), were exposed to the beam of a 1040 nm Yb-based amplified fs-laser system with a pulse length of about 350 fs. With appropriate parameters, the laser exposure resulted in a surface topography with oriented micro-grooves, which, for PC, were covered with nano-ripples. Schwann cell growth on these substrates was inspected after 3 to 5 days of cultivation by means of scanning electron microscopy (SEM). We show that Schwann cells can grow in a certain direction, predetermined by micro-groove or nano-ripple orientation. In contrast, cells cultivated on randomly oriented nanofibers or unstructured surfaces show an omnidirectional growth behavior. This method may be used in the future to produce nerve conduits for the treatment of injuries to the peripheral nervous system. Full article
(This article belongs to the Special Issue Ultrafast and Nonlinear Laser Applications)
Show Figures

Figure 1

14 pages, 1327 KiB  
Article
Exploration of Cytokines That Impact the Therapeutic Efficacy of Mesenchymal Stem Cells in Alzheimer’s Disease
by Herui Wang, Chonglin Zhong, Yi Mi, Guo Li, Chenliang Zhang, Yaoyao Chen, Xin Li, Yongjun Liu and Guangyang Liu
Bioengineering 2025, 12(6), 646; https://doi.org/10.3390/bioengineering12060646 - 12 Jun 2025
Viewed by 535
Abstract
Current therapies for Alzheimer’s disease (AD) includes acetylcholinesterase inhibitors, NMDA receptor antagonists, and amyloid beta (Aβ)/Tau-targeting drugs. While these drugs improve cognitive decline and target the pathological mechanisms, their outcomes still are still in debate. Mesenchymal stem cells (MSCs) offer a regenerative approach [...] Read more.
Current therapies for Alzheimer’s disease (AD) includes acetylcholinesterase inhibitors, NMDA receptor antagonists, and amyloid beta (Aβ)/Tau-targeting drugs. While these drugs improve cognitive decline and target the pathological mechanisms, their outcomes still are still in debate. Mesenchymal stem cells (MSCs) offer a regenerative approach by modulating neuroinflammation and promoting neuroprotection. Although the paracrine of MSCs is efficient in various AD preclinical studies and the exosomes of MSCs have entered clinical trials, the key cytokines driving the efficacy remain unclear. Here, we evaluated human umbilical cord-derived MSCs (hUC-MSCs) and employed gene-silenced MSCs (siHGF-MSCs, siTNFR1-MSCs, siBDNF-MSCs) in APP/PS1 AD mice to investigate specific mechanisms. hUC-MSCs significantly reduced Aβ/Tau pathology and neuroinflammation, with cytokine-specific contributions: silencing HGF predominantly reduced Aβ/Tau clearance, although silencing TNFR1 or BDNF showed modest effects; silencing TNFR1 or BDNF more prominently weakened anti-neuroinflammation, while silencing HGF exerted a weaker influence. All three cytokines partially contributed to oxidative stress reduction and cognitive improvements. Our study highlights MSC-driven AD alleviation as a multifactorial strategy and reveals specific cytokines alleviating different aspects of AD pathology. Full article
(This article belongs to the Special Issue Nerve Regeneration)
Show Figures

Graphical abstract

30 pages, 2629 KiB  
Review
Pericytes and Diabetic Microangiopathies: Tissue Resident Mesenchymal Stem Cells with High Plasticity and Regenerative Capacity
by Zeinab Shirbaghaee, Christine M. Sorenson and Nader Sheibani
Int. J. Mol. Sci. 2025, 26(11), 5333; https://doi.org/10.3390/ijms26115333 - 1 Jun 2025
Viewed by 974
Abstract
Pericytes (PCs), a heterogeneous population of perivascular supporting cells, are critical regulators of vascular stability, angiogenesis, and blood–tissue barrier integrity. Increasing evidence highlights their active role in the pathophysiology of diabetic microangiopathies, including those affecting the retina, kidney, brain, heart, and peripheral nerves. [...] Read more.
Pericytes (PCs), a heterogeneous population of perivascular supporting cells, are critical regulators of vascular stability, angiogenesis, and blood–tissue barrier integrity. Increasing evidence highlights their active role in the pathophysiology of diabetic microangiopathies, including those affecting the retina, kidney, brain, heart, and peripheral nerves. In diabetes, hyperglycemia-induced PC dysfunction is a major contributor to vascular degeneration, impaired tissue repair, and disease progression across multiple organs. Pericytes also share many characteristics with mesenchymal stem cells (MSCs). They exhibit regenerative capacity, immunomodulatory activities, and multipotent capacities. This review explores the emerging role of PCs as tissue resident MSCs, emphasizing their pathophysiological involvement in diabetes complications, and their potential for utilization in regenerative medicine. We also discuss advances in PC-based therapies, tissue engineering strategies, and clinical applications. Thus, PCs are positioned as promising targets for therapeutic intervention and vascular tissue regeneration. Full article
(This article belongs to the Special Issue Diabetes and Metabolic Dysfunction)
Show Figures

Figure 1

Back to TopTop