321. Immunoregulation of Quercetin and Kynurenic Acid on Human Umbilical Cord Mesenchymal Stem Cells Through Gene Expression of Aryl Hydrocarbon Receptor and Interleukin-6 in Hyperglycemic Milieu.
作者: Thi Sam Nguyen.;Thi Thuy Ngan Nguyen.;Thi Phuong Anh Nguyen.;Tran Bao Chau Ha.;Manh Cuong Nguyen.;Syed Shadab Raza.;Vinh Truong Do.;Hoang Ha Chu.
来源: Stem Cells Int. 2025年2025卷6612312页
Mesenchymal stem cells (MSCs) exhibit great promise for treatment applications because of their immunosuppressive properties. The aryl hydrocarbon receptor (AHR), which is a transcription factor that is activated via ligand, has a pivotal role in regulating the immune system and is involved in a range of immune-related disorders. However, hyperglycemia, the defining biochemical hallmark of diabetes, creates a chronically pro-inflammatory microenvironment that impairs the immunoregulatory effects of MSCs. In this study, we explored the potential of kynurenic acid (KYNA) and quercetin, two naturally derived compounds, to modulate the immune response of MSCs through the regulation of AHR signaling under hyperglycemic conditions. We assessed the immunophenotyping and differentiation capacity of cultured human umbilical cord mesenchymal stem cells (hUC-MSCs) in a high-glucose medium and quantified the mRNA expression rate of AHR, CYP1A1, CYP1B1, and IL-6 using real time PCR. Our study is the first to reveal that KYNA and quercetin enhance mRNA expression levels of AHR and CYP1B1, while reducing IL-6 expression in hUC-MSCs, suggesting their potential as immunomodulators. These findings highlight the compounds' promise as drug candidates for immune-mediated diseases through stem cell therapy, particularly due to their modulation of AHR.
322. Isolation and Characterization of Cervical Cancer-Associated Mesenchymal Stem Cells From Primary Tumors Using Explant Culture.
作者: Surbhi Singla.;Sanchita Khurana.;Rashmi Bagga.;Radhika Srinivasan.;Shalmoli Bhattacharyya.
来源: Bio Protoc. 2025年15卷12期e5358页
Cancer-associated mesenchymal stem cells (Ca-MSCs), an integral part of the tumor microenvironment, play a major role in modulating tumor progression; they have been reported to progress as well as inhibit various cancers, including cervical cancer. To understand the exact role of Ca-MSCs in tumor modulation, it is necessary to have an optimized protocol for Ca-MSCs isolation. This work demonstrates the isolation and expansion of a primary culture of cervical cancer-associated MSCs (CCa-MSCs) from the biopsy sample of cervical cancer patients using the explant culture technique. The isolated cells were characterized according to International Society for Cellular Therapy (ISCT) guidelines. Morphological analysis revealed that cells were adherent to the plastic surface and possessed spindle-shaped morphology. Flow cytometry analysis of the cells showed high expression (~98%) for MSC-specific cell surface markers (CD90, CD73, and CD105), negative expression (<0.5%) for endothelial cell marker (CD34) and hematopoietic cell marker (CD45), and negligible expression for HLA-DR, as recommended by ISCT. Further, trilineage differentiation potential analysis of the cells showed their osteogenic and chondrogenic potential and adipogenic differentiation. This standardized protocol will assist in the cultivation of CCa-MSCs and the study of their interactions with tumor cells and other components of the tumor microenvironment. This protocol may be utilized in the establishment of Ca-MSCs from other types of cancers as well. Key features • Isolation and expansion of cervical cancer-associated mesenchymal stem cells (CCa-MSCs) from patient biopsy sample. • Characterization of isolated CCa-MSCs for the presence of MSC-specific cell surface markers and trilineage differentiation potential. • CCa-MSCs can be employed to study the interactions with the tumor cells in the tumor microenvironment. Graphical overview.
323. A Hybrid 2D/3D Approach for Neural Differentiation Into Telencephalic Organoids and Efficient Modulation of FGF8 Signaling.
Human brain development relies on a finely tuned balance between the proliferation and differentiation of neural progenitor cells, followed by the migration, differentiation, and connectivity of post-mitotic neurons with region-specific identities. These processes are orchestrated by gradients of morphogens, such as FGF8. Disruption of this developmental balance can lead to brain malformations, which underlie a range of complex neurodevelopmental disorders, including epilepsy, autism, and intellectual disabilities. Studying the early stages of human brain development, whether under normal or pathological conditions, remains challenging due to ethical and technical limitations inherent to working with human fetal tissue. Recently, human brain organoids have emerged as a powerful in vitro alternative, allowing researchers to model key aspects of early brain development while circumventing many of these constraints. Unlike traditional 2D cultures, where neural progenitors and neurons are grown on flat surfaces, 3D organoids form floating self-organizing aggregates that better replicate the cellular diversity and tissue architecture of the developing brain. However, 3D organoid protocols often suffer from significant variability between batches and individual organoids. Furthermore, few existing protocols directly manipulate key morphogen signaling pathways or provide detailed analyses of the resulting effects on regional brain patterning. • To address these limitations, we developed a hybrid 2D/3D approach for the rapid and efficient induction of telencephalic organoids that recapitulate major steps of anterior brain development. Starting from human induced pluripotent stem cells (hiPSCs), our protocol begins with 2D neural induction using small-molecule inhibitors to achieve fast and homogenous production of neural progenitors (NPs). After dissociation, NPs are reaggregated in Matrigel droplets and cultured in spinning mini-bioreactors, where they self-organize into neural rosettes and neuroepithelial structures, surrounded by differentiating neurons. Activation of the FGF signaling pathway through the controlled addition of FGF8 to the culture medium will modulate regional identity within developing organoids, leading to the formation of distinct co-developing domains within a single organoid. Our protocol combines the speed and reproducibility of 2D induction with the structural and cellular complexity of 3D telencephalic organoids. The ability to manipulate signaling pathways provides an additional opportunity to further increase system complexity, enabling the simultaneous development of multiple distinct brain regions within a single organoid. This versatile system facilitates the study of key cellular and molecular mechanisms driving early human brain development across both telencephalic and non-telencephalic areas. Key features • This protocol builds on the method established by Chambers et al. [1] for generating 2D neural progenitors, followed by dissociation and reaggregation into 3D brain organoids. • For optimal growth and maturation, telencephalic organoids are cultured in spinning mini-bioreactors [2] or on orbital shakers. • The protocol enables the generation of telencephalic neural progenitors in 10 days and produces 3D telencephalic organoids containing neocortical neurons within one month of culture. • Addition of morphogens in the culture medium (example: FGF8) enhances cellular heterogeneity, promoting the emergence of distinct brain domains within a single organoid.
324. Mechanistic insights into adipose-derived stem cells and exosomes in ischemia-reperfusion injury repair: from shared pathways to organ-specific therapeutics.
作者: Jiaqian Si.;Jie Wang.;Hao Dai.;Tuochen Lv.;Songyun Zhao.;Wanying Chen.;Liqun Li.;Siqi Ding.;Yucang He.
来源: Front Cell Dev Biol. 2025年13卷1621289页
Ischemia-reperfusion injury (IRI) has become a significant challenge for clinical treatment due to the complex multi-mechanism pathological cascade response, including oxidative stress, inflammatory bursts, and programmed cell death. Adipose-derived stem cells (ADSCs) and their exosomes (ADSCs-exosomes) are emerging as a breakthrough therapeutic strategy to reverse IRI, owing to their multi-target synergistic effects. This review systematically analyzes the two major repair modes of ADSCs and ADSCs-exosomes: the "common protection" mechanism, which includes anti-inflammatory, anti-oxidative, and anti-apoptotic effects through paracrine regulation of miRNAs targeting the NF-κB/NRF2/β-catenin signaling axis; and precision repair, which is achieved through organ-specific targets, including hepatic mitochondrial dynamics and pyroptosis inhibition, cardiac macrophage polarization and neutrophil clearance, renal anti-fibrosis and erythropoietin (EPO) activation, as well as brain iron death regulation and microglial remodeling. From the perspective of the mechanism interaction network, this paper first proposes a theoretical framework of "multi-organ shared core pathways and dynamic regulation of different targets." It also reviews the translational potential of combined therapeutic strategies based on engineered exosomes delivery systems and biomaterials, emphasizing the optimization of delivery efficiency and functional enhancement to address the bottleneck of clinical applications. The ADSCs-mediated IRI intervention system provides an essential theoretical and technical basis for the development of individualized precision therapies.
325. Current approaches for Usher syndrome disease models and developing therapies.
作者: Fiona K Leith.;Joey Lye.;Derek S Delaney.;Samuel McLenachan.;Fred K Chen.;Marcus D Atlas.;Elaine Y M Wong.
来源: Front Cell Dev Biol. 2025年13卷1547523页
Usher syndrome is a severely debilitating autosomal recessive disorder characterised by congenital or progressive hearing loss, gradual vision loss and in some subtypes, vestibular dysfunction. Much progress has been made in recent years in creating appropriate preclinical models for most subtypes of Usher syndrome to facilitate the development of novel therapies. In this review, we provide an update on new preclinical models of Usher syndrome, with a particular focus on induced pluripotent stem cells and new organoid models. An update on the status of novel therapies is provided, including the development of new genetic therapies using new preclinical models and those currently in clinical trials.
326. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Emerging Therapies for Neurodegenerative Diseases.
作者: Puwen Chen.;Fangsheng Wang.;Baodian Ling.;Yifan Zhu.;Haihong Lin.;Junyun Huang.;Xiaoling Wang.
来源: Int J Nanomedicine. 2025年20卷8547-8565页
Neurodegenerative diseases are a group of chronic diseases characterized by a gradual loss of neurons that worsens over time and dysfunction. These diseases are extremely harmful, not only affecting the physical health of the patients, but also having a serious impact on their quality of life. They mainly include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS), etc. Their pathogenesis is complex, and it is difficult for the existing treatments to effectively slow down the progression of the disease. In recent years, Mesenchymal Stem Cells (MSCs) have received widespread attention for their anti-inflammatory, immunomodulatory and neuroprotective properties. In this context, MSC-derived Extracellular Vesicles (MSC-EVs) have demonstrated unique therapeutic potential as a cell-free therapeutic strategy. MSC-EVs are rich in bioactive substances such as proteins, lipids, mRNAs and miRNAs, which can pass through the blood-brain barrier and be targeted to the diseased area to regulate neuronal survival, synaptic plasticity and neuroinflammatory responses. In addition, compared with stem cell therapy, MSC-EVs have the advantages of low immunogenicity, easy storage and transportation, and avoiding ethical controversies. However, their clinical application still faces challenges: standardized isolation and purification techniques have not been unified, vesicle loading efficiency and targeting need to be further optimized, and long-term safety needs to be systematically evaluated. This review focuses on the role of MSC-EVs in the development of neurological diseases and explores their possible dual roles, both favorable and unfavorable, in the context of neurological diseases. In addition, this review provides a review of current studies on EVs as potential biomarkers for the diagnosis and treatment of neurodegenerative diseases and provides a comprehensive review of the prospects and challenges of MSC-EVs in clinical applications.
327. Aberrant glucose metabolism drives dysfunction of CD4+ T cells in systemic lupus erythematosus and disease flares.
作者: Lu Jin.;Meng Ding.;Shaoxin Cui.;Lin Yang.;Jinwen Zhao.;Jingjing He.;Xiaoping Wang.;Fei Chang.;Xue Liu.;Qun Wang.;Hongtao Jin.;Jun Ma.;Aijing Liu.
来源: Cent Eur J Immunol. 2025年50卷1期13-23页
T cell immuno-metabolic regulation plays a key role in the development of systemic lupus erythematosus (SLE). This study aimed to analyze the role of CD4+ T cell glucose metabolism in SLE development.
328. Comments on: "Long-term efficacy of mesenchymal stem cell treatment for complex perianal fistulas: A systematic review and meta-analysis".
The meta-analysis by Wang et al. aimed to assess the long-term effects of mesenchymal stem cells on complex perianal fistula. The authors concluded that mesenchymal stem cell therapy has a long-term effect on the clinical response of complex perianal fistula and should be widely promoted not only in adults but also in infants and adolescents; however, more research on this topic is needed. We appreciate the authors' hard work, and we also agree with this argument. However, we have several concerns about the study. We think it is necessary to discuss the effect of anti-TNF and immunosuppressive therapy on the effcacy of mesenchymal stem cell treatment for perianal fistula in future trials, in order to optimize treatment strategies in perianal fistula patients and reduce the economic burden of patients. In the future, it will be interesting to assess the safety and feasibility of injection of fibrin glue combined with mesenchymal stem cells in perianal fistula.
329. Stem cell-based therapeutic strategies for liver aging.
作者: Huan Niu.;Yan-Nan Wang.;Yu Ding.;Yu-Qing Lin.;Jian Qin.;Jian-Cheng Wang.
来源: Liver Res. 2025年9卷2期118-131页
Aging is characterized by a gradual deterioration of the physiological integrity of cells, tissues, and organs, resulting in a decrease in the body's physiological functions and an acceleration of the onset of age-related diseases, ultimately leading to death. The aging of the liver, which is a critical metabolic organ, is closely linked to various chronic liver diseases, such as hepatitis, liver fibrosis, and cirrhosis, and it exacerbates their prognosis and is a primary risk factor for their development at all stages. Therefore, a comprehensive understanding of the causes, mechanisms, and potential therapeutic targets associated with liver aging holds significant clinical importance for delaying or potentially reversing liver aging and for treating chronic liver diseases. Stem cells, which are potential anti-aging agents, present a promising and effective alternative for managing liver aging. In this review, we systematically assess the driving factors, characteristics, and underlying mechanisms of liver aging. We then discuss the current status of the use of stem cells to mitigate liver senescence and address related liver diseases. The review reveals that a stem cell-based approach represents a promising therapeutic strategy for combating liver aging and associated diseases.
330. Soluble CCR2-Expressing Mesenchymal Stem Cells Inhibit Osteoarthritis Development and Progression.
作者: Hyun Sik Na.;Seon-Young Lee.;Dong Hwan Lee.;Keun-Hyung Cho.;Seon Ae Kim.;Eun Jeong Go.;A Ram Lee.;Jeong Su Lee.;Yeon Su Lee.;In Gyu Um.;Se Gyeong Han.;Mi-La Cho.;Seok Jung Kim.
来源: Immune Netw. 2025年25卷3期e24页
Many studies of osteoarthritis (OA) have focused on the use of pain-suppressing drugs and stem cell treatments for cartilage repair. In a previous study, we reported the therapeutic effect of soluble C-C chemokine receptor type 2 (sCCR2) gene therapy on OA. Here, we aimed to demonstrate that sCCR2-expressing stem cells exhibits superior efficacy compared to mesenchymal stem cell (MSC) alone. We used monosodium iodoacetate to induce OA in a Wistar rat model for our experiments. Soluble form of CCR2 was transfected into chondrocytes. We analyzed both in vitro and in vivo systems using sCCR2 E3-transfected MSCs (sCEMs). MCP-1 reduced chondrogenesis, whereas sCEMs improved it. Additionally, disease development was suppressed in MCP-1 conditional knockout mice. In the OA rat model, injection of sCEMs showed significant effects with respect to pain control and reduction of joint cartilage inflammation and damage compared with injection of MOCK-MSCs. These findings indicate that sCEMs inhibit MCP-1, reducing pain and OA-induced cartilage damage and inducing chondroprotection. Inhibiting MCP-1/CCR2 signaling has a significant therapeutic effect on OA. Therefore, sCEM may be an effective treatment for OA.
331. Strong Immune Privileges of MSC and Other Nes-GFP+ Progenitors in Bone Marrow of Transgenic Mice.
Immune privileges (IPs) have been demonstrated for several types of quiescent stem cells (SCs) in adult mammalian organisms. Cancer SCs (CSCs) also manifest IPs, which is one of the main concerns in cancer therapy. To elucidate the strength of non-pathological SCs IPs, we implanted bone marrow (BM) of transgenic N-GFPCF1 mice under the renal capsule of non-transgenic mice immunized with enhanced green fluorescent protein (GFP). We show that Nes-GFP+ mesenchymal SCs (MSCs) and other subpopulations of Nes-GFP+ progenitors in mouse BM have strong IPs, since they survive for at least six wk despite targetedly activated against them full immune system. We also demonstrate that MSCs retain their functionality after exposition to such immune pressure by successful retransplantation of the foci into the secondary non-immunized recipients. We suggest that nestin could be a marker for a wide spectrum of quiescent SCs with strong IPs, including CSCs.
332. Injectable hydrogel loaded with exosomes from hypoxic umbilical cord-derived mesenchymal stem cells alleviates intervertebral disc degeneration by reversing nucleus pulposus cell senescence.
作者: Xin Zhao.;Yubo Shi.;Zhen Sun.;Wei Duan.;Le Chang.;Benchi Xu.;Kangwei Lai.;Jingchun Zhang.;Buqi Tian.;Weidong Tao.;Zhenzhou Mi.;Mian Zhang.;Wenjing Yang.;Zhuojing Luo.;Zhengxu Ye.
来源: Regen Biomater. 2025年12卷rbaf039页
Intervertebral disc degeneration is a significant contributor to the development of spinal disorders. Previous studies have shown that the senescence of nucleus pulposus cells can worsen the degradation of intervertebral disks. Therefore, targeting the senescence of nucleus pulposus cells may be a promising therapeutic approach for the treatment of intervertebral disc degeneration. This study investigated the use of exosomes from hypoxic umbilical cord-derived mesenchymal stem cells to reverse nucleus pulposus cells senescence and delay intervertebral disc degeneration progression. MicroRNA sequencing of hypoxic umbilical cord-derived mesenchymal stem cells revealed the presence of functional microRNAs, with the p53 signalling pathway identified as a key factor. To enhance the release time of hypoxic umbilical cord-derived mesenchymal stem cells in vivo, hyaluronic acid methacryloyl hydrogel was used to load hypoxic umbilical cord-derived mesenchymal stem cells and create a sustained-release system. This system effectively repaired the degradation of the extracellular matrix, reversed nucleus pulposus cells senescence and alleviated intervertebral disc degeneration progression in a rat model. Overall, this study highlights the potential of hypoxic umbilical cord-derived mesenchymal stem cells in reducing nucleus pulposus cell senescence and suggests the possibility of combining it with a sustained-release system as a novel therapeutic strategy for intervertebral disc degeneration.
333. Airway epithelial cell chimerism and chronic lung allograft dysfunction associated with ischemia-reperfusion-injury in lung transplantation.
作者: Yasufumi Goda.;Nirmal S Sharma.;Andrew S Potter.;Don Hayes.
来源: Curr Opin Organ Transplant. 2025年
The ischemia-reperfusion injury (IRI) of the bronchial epithelium after lung transplant (LTx) leads to tissue-specific stem cells (TSC) activation, promoting their migration and facilitation of airway remodeling characterized by a chimeric mixture of donor-derived and recipient-derived epithelial cells. This process results in airway epithelial cell chimerism, which we will discuss in this review as having a role in the pathogenesis of chronic lung allograft dysfunction (CLAD) in LTx recipients (LTRs).
334. Injectable scaffolds with a hierarchically porous structure and augmented paracrine activity for minimally invasive precision medicine.
作者: Lin Du.;Hongjian Zhang.;Ziyi Zhao.;Xueru Ma.;Jimin Huang.;Jinzhou Huang.;Chengtie Wu.
来源: Mater Horiz. 2025年
Stem cell-based therapeutic approaches hold great potential for regenerating severe tissue defects, however, current cell transplantation strategies cannot simultaneously achieve minimally invasive injection and structural integrity. Herein, an injectable 3D-bioprinted scaffold encapsulated with two-phase emulsion bioink-engineered mesenchymal stromal cells is proposed, which can serve as a novel versatile platform for efficient stem cell delivery in minimally invasive approaches. The two-phase emulsion bioinks could spontaneously undergo a phase separation process in an aqueous environment to form hierarchically porous structures (macropore size of 1 mm and micropore size of 100-200 µm), which not only facilitated the proliferation and spreading of the encapsulated cells but also endowed the bioprinted scaffolds with shape memory properties for in situ injection. More importantly, the interconnected macro-microporous structures of the 3D extracellular matrix (ECM) microenvironment could provide biophysical cues to obviously enhance the paracrine functions of encapsulated cells, with the enhanced secretion of bioactive factors related to immunomodulation, angiogenesis, and neurogenesis. RNA-seq results showed that ECM-receptor interaction, focal adhesion, and cytoskeleton regulation might participate in mechanotransduction pathways, thereby enhancing cell paracrine functions. In addition, the bioprinted scaffolds could be in situ injected into skin wounds without damaging their inherent porous structure and, thus, effectively promoted neuro-vascularized skin regeneration by inducing angiogenesis, promoting neurogenesis and suppressing the inflammatory response. Taken together, we successfully prepared an injectable scaffold integrating hierarchically porous structures with the augmented paracrine activity of stem cells, which is a promising candidate for tissue regeneration and minimally invasive precision medicine.
335. [Observation of the effect of Yanghe Pingchuan granules on the homing of BMSCs in asthma based on FTO regulation of Notch1 pathway].
作者: Kun Wang.;Haoxiang Fang.;Xiaomei Cao.
来源: Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2025年41卷7期585-592页
Objective To observe the effect of m6A methylation regulation on Notch1 pathway on the homing of BMSCs in asthma, and the intervention study of traditional Chinese medicine compound Yanghe Pingchuan Granules. Methods Rat bone mesenchymal stem cells(BMSC)and bronchial epithelial cells were cocultured. The extracted cells were divided into: bronchial epithelial cell group, asthma bronchial epithelial cell+mesenchymal stem cell co-culture group (co-culture group), co-culture cell+normal serum group, coculture cell+serum containing optimal drug group, siRNA FTO+normal serum group, siRNA FTO-NC+normal serum group, and siRNA FTO+serum containing optimal drug group. The vitality and cell cycle changes of co-cultured cells were detected. The level and markers of homing BMSC were detected by immunofluorescence staining. The expression of Notch1 pathway related genes were detected by qRT-PCR. The expression of Notch1 pathway related proteins were detected by Western blot. Results Compared with bronchial epithelial cell group, the co-cultured cell group showed an increase in the homing level of BMSCs and the expression of C-X-C motif chemokine receptor 4 (CXCR4), stromal cell-derived factor 1 (SDF-1), Notch1, transcription factor recombination signal binding protein-J (RBP-J), and hairy enhancer of split 1 (Hes1) proteins. Compared with the co-cultured cell group and co-cultured cell+normal serum group, the co-cultured cell+serum containing optimal drug group showed an increase in the homing level of BMSCs and the expressions of CXCR4 and SDF-1, while the protein and mRNA levels of Notch1 and Hes1 decreased. Compared with the siRNA FTO-NC+normal serum group, the siRNA FTO+normal serum group showed an increase in the levels of Notch1, activated Notch1, RBP-J, Hes1 protein, and cell viability, while the level of homing BMSC decreased. Compared with siRNA FTO+normal serum group, the levels of Notch1, RBP-J mRNA, activated Notch1, and Hes1 protein decreased, while the level of homing BMSCs increased in siRNA FTO+serum containing optimal drug group. The levels of Notch1, RBP-J, and Hes1 mRNA were reduced in the co-cultured cells+serum containing optimal drug group. Compared with siRNA FTO+serum containing optimal drug group, the expressions of Notch1, activated Notch1, RBP-J, Hes1 protein and cell viability decreased, while the level of homing BMSCs increased in the co-cultured cells+serum containing optimal drug group. Conclusion Yanghe Pingchuan Granules may promote the homing of BMSCs in asthma and alleviate asthma inflammation by upregulating the expression of FTO and inhibiting the expression of downstream genes in the Notch1 signaling pathway.
336. Umbilical Cord-Mesenchymal Stromal Cell-Derived Extracellular Vesicles Target the Liver to Improve Neurovascular Health in Type 2 Diabetes With Non-Alcoholic Fatty Liver Disease.
作者: Minghao Du.;Hao Yang.;Jinyun Niu.;Jing Huang.;Lihong Wang.;Junxiu Xi.;Panpan Meng.;Zhiyong Liu.;Guaiguai Ma.;Jiani Li.;Xiaoyan Liu.;Liang Guo.;Mingjun Hu.;Zhufang Tian.;Bin Liu.;Weiping Liu.;Ashok K Shetty.;Shengxi Wu.;Andrius Baskys.;Qianfa Long.
来源: J Extracell Vesicles. 2025年14卷7期e70125页
Type 2 diabetes mellitus (T2DM) combined with non-alcoholic fatty liver disease (NAFLD) exacerbates metabolic dysregulation and neurovascular complications, presenting significant therapeutic challenges. We demonstrate, using SPECT/CT imaging, that extracellular vesicles (EVs) from mesenchymal stromal cells (MSCs) predominantly accumulate in the liver, where they deliver miR-31-5p to suppress platelet-derived growth factor B (PDGFB) produced by hepatic macrophages. This intervention impedes NAFLD progression and establishes a mechanistic link between liver repair and neurovascular improvement. Specifically, single-nucleus RNA sequencing reveals that PDGFB suppression enhances hippocampal pericyte recovery via the PDGFB-PDGFRβ axis and orchestrates the activation of growth differentiation factor 11 (GDF11), thus promoting neuroplasticity. Furthermore, AAV injections indicate that hepatic PDGFB modulation recalibrates transthyretin (TTR) dynamics, thereby restoring its neuroprotective functions and preventing its pathological deposition in the brain. These findings position MSC-EVs as a transformative therapeutic platform that leverages the liver-brain axis to address the intertwined metabolic and neurovascular complications of T2DM, offering a promising avenue for clinical translation.
337. Methylation levels in keratinocyte basal cells reflect donor age and associate with altered cellular proliferation pathways.
To determine if epigenomic and mRNA associations with donor age are present in basal keratinocytes in vitro.
338. Current Anti-Myeloma Chimeric Antigen Receptor-T Cells: Novel Targets and Methods.
Multiple myeloma (MM) treatment becomes a major challenge once triple-class or penta-refractoriness develops. Emerging immunotherapies, including bispecific antibodies or chimeric antigen receptor (CAR)-T cell therapy, are promising options for such patients. However, the requirement for specialized expertise and staff under stringent manufacturing conditions results in high costs and restricted production. This article explores the manufacturing and clinical application of CAR T-cells in MM, highlighting their potential, limitations, and strategies to enhance efficacy. CAR-T can be manufactured by pharmaceutical companies or accredited academic centers authorized to produce and market gene-edited cellular products. This process includes sequential steps: T cell apheresis from the patient, selection of the cells, activation, gene transfer, expansion of the produced cells, cryopreservation, and reinfusion of the cells into a lymphodepleted patient. While CD3+ T cells are typically employed for CAR-T production in clinical studies, studies have demonstrated the potential advantages of specific T cell subgroups, such as naive, central memory, and memory stem cells, in enhancing efficacy. Following T cell harvesting, the subsequent phase involves genetic modification. CAR-T cells are frequently produced by applying viral vectors such as γ-retrovirus or lentivirus. Although viral vectors are commonly used, non-viral methods-including CRISPR/Cas9 and integrative mRNA transfection methods produced by transposons-are also employed. Five different CAR-T cell generations have been developed. The myeloma-specific targets B-cell maturation antigen (BCMA), signaling lymphocyte activation molecular family 7, and G protein-coupled receptor class C group 5 member D are the most extensively studied in clinical trials. Emerging CAR-T cell targets under investigation include CD138, CD19, kappa light chain, CD56, NY-ESO-1, CD70, TACI, and natural killer G2D. In 2021, idecabtagene vicleucel, a BCMA-targeting agent, became the first CAR-T therapy approved for relapsed/refractory MM, marking a significant milestone in MM treatment. Subsequently, ciltacabtagene autoleucel has also been approved. However, CAR-T resistance is an emerging issue. Resistance mechanisms include T cell exhaustion, antigen escape (loss of BCMA), and tumor microenvironment-related inhibitors. To address these challenges, strategies such as BCMA non-targeted or dual-targeted CAR-T, memory T cells, humanized CAR-T, and rapidly manufactured PHE885 cells have been developed. To enhance specificity, ongoing investigations include bicistronic CAR/co-stimulator receptors, formation of memory-phenotype T cells, combination with immunomodulators or checkpoint inhibitors, armored CAR-T cells, cancer-associated fibroblast inhibitors, and CAR approaches that inhibit exhaustion signals. In conclusion, studies are exploring the use of CAR-T at an earlier stage, including at diagnosis, with an aim to replace ASCT. CAR-T has introduced a new dimension to MM treatment; however, limited efficacy in high-risk MM and the emergence of resistance to CAR-T remain key challenges to be addressed.
339. Caenorhabditis elegans germline flux pulls its stem-cell niche cells into shape.
Morphogenesis is the process by which tissues and organs acquire their unique shapes and functions. Several cellular events, including cell shape changes, cell movement, cell proliferation, apoptosis, and others, act in concert to sculpt an organ. While these cellular behaviors are well-recognized to be essential for morphogenesis, whether organ-scale dynamics influence these cellular processes has remained unresolved. In a recent study published in Development, Tolkin et al. (2024) investigated the mechanisms underlying the dramatic morphogenesis of the distal tip cell (DTC), a leader cell and germline stem-cell niche in the Caenorhabditis elegans gonad (Byrd et al. 2014; Lee et al. 2016). They uncovered a novel 'hitch-and-tow' mechanism whereby germline flux drives a complex change in DTC shape. This work sheds light on how mechanical forces at the organ level influence cellular morphogenesis.
340. Stem Cell Therapy and Models for Autism Spectrum Disorder: Insights and Research.
作者: Yulong Liu.;Yi Luo.;Jiayin Liu.;Meifeng Gong.;Meiling Xia.;Xiaotang Fan.
来源: Curr Neuropharmacol. 2025年
Autism Spectrum Disorders (ASD) are complex neurodevelopmental conditions characterized by impaired social communication, repetitive behavior patterns, and atypical sensory perception. The Autism and Developmental Disabilities Monitoring Network reports that approximately 1 in 36 children are diagnosed with ASD, highlighting the increasing prevalence and the pressing need for innovative treatment approaches. Medications commonly used in ASD primarily aim to manage associated symptoms, as there are currently no FDA-approved medications specifically for treating ASD core symptoms. Stem cells have demonstrated significant potential in cell-based therapies for ASD and have been utilized in in vitro models to investigate the pathogenesis of the condition. This review focuses on the recent advancements in stem cell-based transplantation in animal models of ASD, aiming to explore the improvement of ASD symptoms and the underlying mechanisms involved. It also discussed the application of stem cell-based transplantation in pediatric and adolescent populations with ASD to evaluate treatment efficacy and potential preventive strategies. Furthermore, recent efforts are addressed in developing stem cell-based models for both syndromic and non-syndromic forms of ASD, emphasizing studies that utilize cerebral organoids for modeling ASD, which facilitate the exploration of disease mechanisms within a tissue-like environment.
|