Sign in to use this feature.

Years

Between: -

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (332)

Search Parameters:
Journal = Biomedicines
Section = Gene and Cell Therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 1282 KiB  
Systematic Review
Actinic Cheilitis: A Systematic Review and Meta-Analysis of Interventions, Treatment Outcomes, and Adverse Events
by Matthäus Al-Fartwsi, Anne Petzold, Theresa Steeb, Lina Amin Djawher, Anja Wessely, Anett Leppert, Carola Berking and Markus V. Heppt
Biomedicines 2025, 13(8), 1896; https://doi.org/10.3390/biomedicines13081896 - 4 Aug 2025
Abstract
Introduction: Actinic cheilitis (AC) is a common precancerous condition affecting the lips, primarily caused by prolonged ultraviolet radiation exposure. Various treatment options are available. However, the optimal treatment approach remains a subject of debate. Objective: To summarize and compare practice-relevant interventions for AC. [...] Read more.
Introduction: Actinic cheilitis (AC) is a common precancerous condition affecting the lips, primarily caused by prolonged ultraviolet radiation exposure. Various treatment options are available. However, the optimal treatment approach remains a subject of debate. Objective: To summarize and compare practice-relevant interventions for AC. Materials and Methods: A pre-defined protocol was registered in PROSPERO (CRD42021225182). Systematic searches in Medline, Embase, and Central, along with manual trial register searches, identified studies reporting participant clearance rates (PCR) or recurrence rates (PRR). Quality assessment for randomized controlled trials (RCTs) was conducted using the Cochrane Risk of Bias tool 2. Uncontrolled studies were evaluated using the tool developed by the National Heart, Lung, and Blood Institute. The generalized linear mixed model was used to pool proportions for uncontrolled studies. A pairwise meta-analysis for RCTs was applied, using the odds ratio (OR) as the effect estimate and the GRADE approach to evaluate the quality of the evidence. Adverse events were analyzed qualitatively. Results: A comprehensive inclusion of 36 studies facilitated an evaluation of 614 participants for PCR, and 430 patients for PRR. Diclofenac showed the lowest PCR (0.53, 95% confidence interval (CI) [0.41; 0.66]), while CO2 laser showed the highest PCR (0.97, 95% CI [0.90; 0.99]). For PRR, Er:YAG laser showed the highest rates (0.14, 95% CI [0.08; 0.21]), and imiquimod the lowest (0.00, 95% CI [0.00; 0.06]). In a pairwise meta-analysis, the OR indicated a lower recurrence rate for Er:YAG ablative fractional laser (AFL)-primed methyl-aminolevulinate photodynamic therapy (MAL-PDT) (Er:YAG AFL-PDT) compared to methyl-aminolevulinate photodynamic therapy (MAL-PDT) alone (OR = 0.22, 95% CI [0.06; 0.82]). The CO2 laser showed fewer local side effects than the Er:YAG laser, while PDTs caused more skin reactions. Due to qualitative data, comparability was limited, highlighting the need for individualized treatment. Conclusions: This study provides a complete and up-to-date evidence synthesis of practice-relevant interventions for AC, identifying the CO2 laser as the most effective treatment and regarding PCR and imiquimod as most effective concerning PRR. Full article
(This article belongs to the Special Issue Skin Diseases and Cell Therapy)
Show Figures

Figure 1

21 pages, 328 KiB  
Review
Adjuvant Immunotherapy in Stage IIB/IIC Melanoma: Current Evidence and Future Directions
by Ivana Prkačin, Ana Brkić, Nives Pondeljak, Mislav Mokos, Klara Gaćina and Mirna Šitum
Biomedicines 2025, 13(8), 1894; https://doi.org/10.3390/biomedicines13081894 - 4 Aug 2025
Viewed by 51
Abstract
Background: Patients with resected stage IIB and IIC melanoma are at high risk of recurrence and distant metastasis, despite surgical treatment. The recent emergence of immune checkpoint inhibitors (ICIs) has led to their evaluation in the adjuvant setting for early-stage disease. This [...] Read more.
Background: Patients with resected stage IIB and IIC melanoma are at high risk of recurrence and distant metastasis, despite surgical treatment. The recent emergence of immune checkpoint inhibitors (ICIs) has led to their evaluation in the adjuvant setting for early-stage disease. This review aims to synthesize current evidence regarding adjuvant immunotherapy for stage IIB/IIC melanoma, explore emerging strategies, and highlight key challenges and future directions. Methods: We conducted a comprehensive literature review of randomized clinical trials, observational studies, and relevant mechanistic and biomarker research on adjuvant therapy in stage IIB/IIC melanoma. Particular focus was placed on pivotal trials evaluating PD-1 inhibitors (KEYNOTE-716 and CheckMate 76K), novel vaccine and targeted therapy trials, mechanisms of resistance, immune-related toxicity, and biomarker development. Results: KEYNOTE-716 and CheckMate 76K demonstrated significant improvements in recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) with pembrolizumab and nivolumab, respectively, compared to placebo. However, no definitive overall survival benefit has yet been shown. Adjuvant immunotherapy is linked to immune-related adverse events, including permanent endocrinopathies. Emerging personalized approaches, such as circulating tumor DNA monitoring and gene expression profiling, may enhance patient selection, but remain investigational. Conclusions: Adjuvant PD-1 blockade offers clear RFS benefits in high-risk stage II melanoma, but optimal patient selection remains challenging, due to uncertain overall survival benefit and toxicity concerns. Future trials should integrate biomarker-driven approaches to refine therapeutic decisions and minimize overtreatment. Full article
(This article belongs to the Section Gene and Cell Therapy)
22 pages, 6395 KiB  
Article
Investigation of Novel Therapeutic Targets for Rheumatoid Arthritis Through Human Plasma Proteome
by Hong Wang, Chengyi Huang, Kangkang Huang, Tingkui Wu and Hao Liu
Biomedicines 2025, 13(8), 1841; https://doi.org/10.3390/biomedicines13081841 - 29 Jul 2025
Viewed by 361
Abstract
Background: Rheumatoid arthritis (RA) is an autoimmune disease that remains incurable. An increasing number of proteomic genome-wide association studies (GWASs) are emerging, offering immense potential for identifying novel therapeutic targets for diseases. This study aims to identify potential therapeutic targets for RA [...] Read more.
Background: Rheumatoid arthritis (RA) is an autoimmune disease that remains incurable. An increasing number of proteomic genome-wide association studies (GWASs) are emerging, offering immense potential for identifying novel therapeutic targets for diseases. This study aims to identify potential therapeutic targets for RA based on human plasma proteome. Methods: Protein quantitative trait loci were extracted and integrated from eight large-scale proteomic GWASs. Proteome-wide Mendelian randomization (Pro-MR) was performed to prioritize proteins causally associated with RA. Further validation of the reliability and stratification of prioritized proteins was performed using MR meta-analysis, colocalization, and transcriptome-wide summary-data-based MR. Subsequently, prioritized proteins were characterized through protein–protein interaction and enrichment analyses, pleiotropy assessment, genetically engineered mouse models, cell-type-specific expression analysis, and druggability evaluation. Phenotypic expansion analyses were also conducted to explore the effects of the prioritized proteins on phenotypes such as endocrine disorders, cardiovascular diseases, and other immune-related diseases. Results: Pro-MR prioritized 32 unique proteins associated with RA risk. After validation, prioritized proteins were stratified into four reliability tiers. Prioritized proteins showed interactions with established RA drug targets and were enriched in an immune-related functional profile. Four trans-associated proteins exhibited vertical or horizontal pleiotropy with specific genes or proteins. Genetically engineered mouse models for 18 prioritized protein-coding genes displayed abnormal immune phenotypes. Single-cell RNA sequencing data were used to validate the enriched expression of several prioritized proteins in specific synovial cell types. Nine prioritized proteins were identified as targets of existing drugs in clinical trials or were already approved. Further phenome-wide MR and mediation analyses revealed the effects and potential mediating roles of some prioritized proteins on other phenotypes. Conclusions: This study identified 32 plasma proteins as potential therapeutic targets for RA, expanding the prospects for drug discovery and deepening insights into RA pathogenesis. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

27 pages, 1201 KiB  
Review
Non-Viral Therapy in COVID-19: Where Are We Standing? How Our Experience with COVID May Help Us Develop Cell Therapies for Long COVID Patients
by Aitor Gonzaga, Gema Martinez-Navarrete, Loreto Macia, Marga Anton-Bonete, Gladys Cahuana, Juan R. Tejedo, Vanessa Zorrilla-Muñoz, Eduardo Fernandez-Jover, Etelvina Andreu, Cristina Eguizabal, Antonio Pérez-Martínez, Carlos Solano, Luis Manuel Hernández-Blasco and Bernat Soria
Biomedicines 2025, 13(8), 1801; https://doi.org/10.3390/biomedicines13081801 - 23 Jul 2025
Viewed by 457
Abstract
Objectives: COVID-19, caused by the SARS-CoV-2 virus, has infected over 777 million individuals and led to approximately 7 million deaths worldwide. Despite significant efforts to develop effective therapies, treatment remains largely supportive, especially for severe complications like acute respiratory distress syndrome (ARDS). [...] Read more.
Objectives: COVID-19, caused by the SARS-CoV-2 virus, has infected over 777 million individuals and led to approximately 7 million deaths worldwide. Despite significant efforts to develop effective therapies, treatment remains largely supportive, especially for severe complications like acute respiratory distress syndrome (ARDS). Numerous compounds from diverse pharmacological classes are currently undergoing preclinical and clinical evaluation, targeting both the virus and the host immune response. Methods: Despite the large number of articles published and after a preliminary attempt was published, we discarded the option of a systematic review. Instead, we have done a description of therapies with these results and a tentative mechanism of action. Results: Preliminary studies and early-phase clinical trials have demonstrated the potential of Mesenchymal Stem Cells (MSCs) in mitigating severe lung damage in COVID-19 patients. Previous research has shown MSCs to be effective in treating various pulmonary conditions, including acute lung injury, idiopathic pulmonary fibrosis, ARDS, asthma, chronic obstructive pulmonary disease, and lung cancer. Their ability to reduce inflammation and promote tissue repair supports their potential role in managing COVID-19-related complications. This review demonstrates the utility of MSCs in the acute phase of COVID-19 and postulates the etiopathogenic role of mitochondria in Long-COVID. Even more, their combination with other therapies is also analyzed. Conclusions: While the therapeutic application of MSCs in COVID-19 is still in early stages, emerging evidence suggests promising outcomes. As research advances, MSCs may become an integral part of treatment strategies for severe COVID-19, particularly in addressing immune-related lung injury and promoting recovery. However, a full pathogenic mechanism may explain or unify the complexity of signs and symptoms of Long COVID and Post-Acute Sequelae (PASC). Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

16 pages, 4308 KiB  
Article
Single-Cell Transcriptomic Analysis of Different Liver Fibrosis Models: Elucidating Molecular Distinctions and Commonalities
by Guofei Deng, Xiaomei Liang, Yuxi Pan, Yusheng Luo, Zizhen Luo, Shaoxuan He, Shuai Huang, Zhaopeng Chen, Jiancheng Wang and Shuo Fang
Biomedicines 2025, 13(8), 1788; https://doi.org/10.3390/biomedicines13081788 - 22 Jul 2025
Viewed by 351
Abstract
Background: Liver fibrosis, a consequence of various chronic liver diseases, is characterized by excessive accumulation of extracellular matrix (ECM), leading to impaired liver function and potentially progressing to cirrhosis or hepatocellular carcinoma. The molecular mechanisms underlying liver fibrosis are complex and not [...] Read more.
Background: Liver fibrosis, a consequence of various chronic liver diseases, is characterized by excessive accumulation of extracellular matrix (ECM), leading to impaired liver function and potentially progressing to cirrhosis or hepatocellular carcinoma. The molecular mechanisms underlying liver fibrosis are complex and not fully understood. In vivo experiments are essential for studying the molecular mechanisms of the disease. However, the diverse principles behind mouse modeling techniques for liver fibrosis can complicate the elucidation of specific fibrotic mechanisms. Methods: Five distinct liver fibrosis models were utilized: CONTROL, NASH (non-alcoholic steatohepatitis), BDL (bile duct ligation), TAA (thioacetamide), and CCl4 (carbon tetrachloride). Patents for these drugs were reviewed using Patentscope® and Worldwide Espacenet®. ScRNA-seq was performed to analyze and compare the cellular and molecular differences in these models. Results: The analysis revealed that, particularly in the drug-induced fibrosis models, hepatic stellate cells (HSCs), Kupffer cells, and T-cell subsets exhibit distinct regulatory patterns and dynamic remodeling processes across different liver fibrosis models. These findings highlight the heterogeneity of immune responses and extracellular matrix (ECM) remodeling in various models, providing important insights into the complex mechanisms underlying liver fibrosis. Conclusions: The study enhances our understanding of liver fibrosis development and provides valuable insights for selecting the most representative animal models in future research. This comprehensive analysis underscores the importance of model-specific immune responses and ECM remodeling in liver fibrosis. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

19 pages, 1083 KiB  
Systematic Review
Revolutionizing Allogeneic Graft Tolerance Through Chimeric Antigen Receptor-T Regulatory Cells
by Alvin Man Lung Chan, Rajalingham Sakthiswary and Yogeswaran Lokanathan
Biomedicines 2025, 13(7), 1757; https://doi.org/10.3390/biomedicines13071757 - 18 Jul 2025
Viewed by 553
Abstract
Background/Objectives: Organ transplantation is a life-saving intervention for patients with terminal organ failure, but long-term success is hindered by graft rejection and dependence on lifelong immunosuppressants. These drugs pose risks such as opportunistic infections and malignancies. Chimeric antigen receptor (CAR) technology, originally [...] Read more.
Background/Objectives: Organ transplantation is a life-saving intervention for patients with terminal organ failure, but long-term success is hindered by graft rejection and dependence on lifelong immunosuppressants. These drugs pose risks such as opportunistic infections and malignancies. Chimeric antigen receptor (CAR) technology, originally developed for cancer immunotherapy, has been adapted to regulatory T cells (Tregs) to enhance their antigen-specific immunosuppressive function. This systematic review evaluates the preclinical development of CAR-Tregs in promoting graft tolerance and suppressing graft-versus-host disease (GvHD). Methods: A systematic review following PROSPERO guidelines (CRD420251073207) was conducted across PubMed, Scopus, and Web of Science for studies published from 2015 to 2024. After screening 105 articles, 17 studies involving CAR-Tregs in preclinical or in vivo transplant or GvHD models were included. Results: CAR-Tregs exhibited superior graft-protective properties compared to unmodified or polyclonal Tregs. HLA-A2-specific CAR-Tregs consistently improved graft survival, reduced inflammatory cytokines, and suppressed immune cell infiltration across skin, heart, and pancreatic islet transplant models. The inclusion of CD28 as a co-stimulatory domain enhanced Treg function and FOXP3 expression. However, challenges such as Treg exhaustion, tonic signaling, and reduced in vivo persistence were noted. Some studies reported synergistic effects when CAR-Tregs were combined with immunosuppressants like rapamycin or tacrolimus. Conclusions: CAR-Tregs offer a promising strategy for inducing targeted immunosuppression in allogeneic transplantation. While preclinical findings are encouraging, further work is needed to optimize CAR design, ensure in vivo stability, and establish clinical-scale manufacturing before translation to human trials. Full article
(This article belongs to the Special Issue Advances in CAR-T Cell Therapy)
Show Figures

Figure 1

26 pages, 6060 KiB  
Article
Identification Exploring the Mechanism and Clinical Validation of Mitochondrial Dynamics-Related Genes in Membranous Nephropathy Based on Mendelian Randomization Study and Bioinformatics Analysis
by Qiuyuan Shao, Nan Li, Huimin Qiu, Min Zhao, Chunming Jiang and Cheng Wan
Biomedicines 2025, 13(6), 1489; https://doi.org/10.3390/biomedicines13061489 - 17 Jun 2025
Viewed by 513
Abstract
Background: Membranous nephropathy (MN), a prevalent glomerular disorder, remains poorly understood in terms of its association with mitochondrial dynamics (MD). This study investigated the mechanistic involvement of mitochondrial dynamics-related genes (MDGs) in the pathogenesis of MN. Methods: Comprehensive bioinformatics analyses—encompassing Mendelian randomization, machine-learning [...] Read more.
Background: Membranous nephropathy (MN), a prevalent glomerular disorder, remains poorly understood in terms of its association with mitochondrial dynamics (MD). This study investigated the mechanistic involvement of mitochondrial dynamics-related genes (MDGs) in the pathogenesis of MN. Methods: Comprehensive bioinformatics analyses—encompassing Mendelian randomization, machine-learning algorithms, and single-cell RNA sequencing (scRNA-seq)—were employed to interrogate transcriptomic datasets (GSE200828, GSE73953, and GSE241302). Core MDGs were further validated using reverse-transcription quantitative polymerase chain reaction (RT-qPCR). Results: Four key MDGs—RTTN, MYO9A, USP40, and NFKBIZ—emerged as critical determinants, predominantly enriched in olfactory transduction pathways. A nomogram model exhibited exceptional diagnostic performance (area under the curve [AUC] = 1). Seventeen immune cell subsets, including regulatory T cells and activated dendritic cells, demonstrated significant differential infiltration in MN. Regulatory network analyses revealed ATF2 co-regulation mediated by RTTN and MYO9A, along with RTTN-driven modulation of ELOA-AS1 via hsa-mir-431-5p. scRNA-seq analysis identified mesenchymal–epithelial transitioning cells as key contributors, with pseudotime trajectory mapping indicating distinct temporal expression profiles: NFKBIZ (initial upregulation followed by decline), USP40 (gradual fluctuation), and RTTN (persistently low expression). RT-qPCR results corroborated a significant downregulation of all four genes in MN samples compared to controls (p < 0.05). Conclusions: These findings elucidate the molecular underpinnings of MDG-mediated mechanisms in MN, revealing novel diagnostic biomarkers and therapeutic targets. The data underscore the interplay between mitochondrial dynamics and immune dysregulation in MN progression, providing a foundation for precision medicine strategies. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

13 pages, 401 KiB  
Article
MAPT Subhaplotypes in Different Progressive Supranuclear Palsy Phenotypes
by Monica Gagliardi, Radha Procopio, Alessia Felicetti, Grazia Annesi, Mariagrazia Talarico, Basilio Vescio, Aldo Quattrone and Andrea Quattrone
Biomedicines 2025, 13(6), 1405; https://doi.org/10.3390/biomedicines13061405 - 7 Jun 2025
Viewed by 544
Abstract
Background: Progressive Supranuclear Palsy (PSP) is a rare neurodegenerative disorder characterized by abnormal tau protein aggregation. The MAPT gene encodes for tau protein. The MAPT locus harbors two major haplotypes, H1 and H2, with H1 and its subhaplotypes being associated with an increased [...] Read more.
Background: Progressive Supranuclear Palsy (PSP) is a rare neurodegenerative disorder characterized by abnormal tau protein aggregation. The MAPT gene encodes for tau protein. The MAPT locus harbors two major haplotypes, H1 and H2, with H1 and its subhaplotypes being associated with an increased risk of PSP. Methods: In this study, we genotyped rs8070723 in a cohort of 73 PSP patients, including 47 PSP Richardson Syndrome (PSP-RS) and 27 PSP variants (vPSP), and 93 age-matched healthy controls (HC) from Southern Italy. Results: Haplotype analysis identified H1 and H2 haplotypes that conferred a risk (OR, 2.620; 95% CI, 1.399–5.140; p = 0.0035) and a protective effect (OR, 0.370; 95% CI, 0.196–0.695; p = 0.0015), respectively. In addition, we genotyped five MAPT variants (rs1467967, rs242557, rs3785883, rs2471738, and rs7521) that, together with rs8070723, defined H1 subhaplotypes. We identified 18 distinct MAPT H1 subhaplotypes, among which H1j displayed a nominally significant reduced risk of PSP (OR, 0.201; 95% CI, 0.044–0.915; p = 0.0265). Conclusions: These findings reinforce the role of MAPT genetic variation in PSP pathogenesis and highlight the potential impact of haplotype diversity on disease susceptibility. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

23 pages, 7919 KiB  
Article
Bone Marrow-Derived Inducible Microglia-like Cells Promote Recovery of Chronic Ischemic Stroke Through Modulating Neuroinflammation in Mice
by Bach Ngoc Nguyen, Tomoaki Kitamura, Shuhei Kobashi, Makoto Urushitani and Tomoya Terashima
Biomedicines 2025, 13(6), 1347; https://doi.org/10.3390/biomedicines13061347 - 30 May 2025
Viewed by 601
Abstract
Background: Chronic ischemic stroke presents a significant challenge in neurology, with limited therapeutic options available for long-term recovery. During cerebral infarction, anti-inflammatory phenotype microglia/macrophages produce anti-inflammatory cytokines and neurotrophic factors that facilitate the process of brain repair. However, obtaining sufficient anti-inflammatory microglia/macrophages from [...] Read more.
Background: Chronic ischemic stroke presents a significant challenge in neurology, with limited therapeutic options available for long-term recovery. During cerebral infarction, anti-inflammatory phenotype microglia/macrophages produce anti-inflammatory cytokines and neurotrophic factors that facilitate the process of brain repair. However, obtaining sufficient anti-inflammatory microglia/macrophages from the human central nervous system is challenging. Bone marrow-derived inducible microglia-like cells (BM-iMGs) with an anti-inflammatory microglial phenotype were explored to induce neuroprotective properties. Here, we transplanted BM-iMGs into the brain of middle cerebral artery occlusion (MCAO) model male mice to explore their potential for treating chronic ischemic stroke. Methods: Bone marrow-derived mononuclear cells (BM-MNCs) were isolated from green fluorescent protein mice and incubated with granulocyte–macrophage colony-stimulating factor (GM-CSF) and IL-4 to induce BM-iMGs with an anti-inflammatory phenotype. BM-iMGs were transplanted into the brains of mice on day 14 after MCAO, and behavioral tests, histology, cerebral blood flow, and gene expression were evaluated. Results: An intracranial injection of BM-iMGs promoted neurobehavioral recovery, reduced neuronal cell loss, suppressed neuroinflammatory astrocytic and microglial responses in the brain, and increased cortical surface cerebral blood flow in MCAO mice. Furthermore, neuroprotective genes were upregulated, whereas proinflammatory genes were downregulated. Conclusions: The intracranial injection of BM-iMG cells shows significant potential as a novel therapy for chronic ischemic stroke. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Graphical abstract

18 pages, 5572 KiB  
Article
Identification of Biomarkers Co-Associated with Lactylation and Acetylation in Systemic Lupus Erythematosus
by Zhanyan Gao, Yang Feng, Chenghui Zheng, Fei Li, Zhan Sun, Mengmeng Xiang, Junrong Zhu, Mingyu Chu, Jinhua Xu and Jun Liang
Biomedicines 2025, 13(6), 1274; https://doi.org/10.3390/biomedicines13061274 - 22 May 2025
Viewed by 632
Abstract
Background: Systemic lupus erythematosus (SLE) is an immune-mediated disease with widespread involvement, and its pathogenesis remains incompletely understood. Recent studies suggest that modifications such as acetylation and lactylation play crucial roles in SLE progression, with potential interrelationships between them. This study aimed to [...] Read more.
Background: Systemic lupus erythematosus (SLE) is an immune-mediated disease with widespread involvement, and its pathogenesis remains incompletely understood. Recent studies suggest that modifications such as acetylation and lactylation play crucial roles in SLE progression, with potential interrelationships between them. This study aimed to identify biomarker genes co-associated with both lactylation and acetylation and to explore their potential mechanisms in SLE pathogenesis. Methods: Microarray data from peripheral blood mononuclear cells (PBMCs) of SLE patients and healthy controls were obtained from the Gene Expression Omnibus (GEO) database. In the training dataset (GSE81622), differential expression analysis was performed to compare SLE samples with healthy controls. Lactate- and acetylation-related genes were used to identify differentially expressed lactate-related genes (LR-DEGs) and acetylation-related genes (AR-DEGs). Genes co-associated with both lactylation and acetylation were further examined. LASSO regression, support vector machine recursive feature elimination (SVM-RFE), and ROC curve analysis were used to identify hub genes. Immune infiltration analysis and a clinical nomogram model were developed for accurate diagnosis and treatment prediction. qPCR was used to validate the hub genes. Results: A total of 1181 differentially expressed genes (DEGs) were identified between SLE and healthy groups. Of these, 33 LR-DEGs and 28 AR-DEGs were identified. Seven genes were found to be co-associated with both lactylation and acetylation. Using LASSO and SVM-RFE, two hub genes, CDCA5 and MCTS1, were identified and validated in the GSE24706 dataset. ROC curve analysis and clinical nomogram revealed significant associations of these biomarkers with SLE pathogenesis. Conclusions: Our study identifies CDCA5 and MCTS1 as potential biomarkers for SLE, potentially influencing its pathogenesis through histone lactylation and acetylation. Experimental validation confirmed their differential expression between SLE patients and healthy controls. These findings underscore the role of epigenetic modifications in SLE, offering new insights into its regulatory mechanisms and immune interactions. Full article
Show Figures

Figure 1

15 pages, 3838 KiB  
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 575
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

25 pages, 840 KiB  
Review
Stem Cell Therapy for Myocardial Infarction Recovery: Advances, Challenges, and Future Directions
by Nicholas T. Le, Matthew W. Dunleavy, William Zhou, Sumrithbir S. Bhatia, Rebecca D. Kumar, Suyin T. Woo, Gonzalo Ramirez-Pulido, Kaushik S. Ramakrishnan and Ahmed H. El-Hashash
Biomedicines 2025, 13(5), 1209; https://doi.org/10.3390/biomedicines13051209 - 16 May 2025
Cited by 1 | Viewed by 1891
Abstract
Myocardial infarction (MI) is a leading cause of morbidity worldwide, resulting from ischemic damage and necrosis to cardiomyocytes. While the standard treatment regimen for MI can be successful in restoring coronary perfusion, it typically does not resolve myocardial damage, which can leave patients [...] Read more.
Myocardial infarction (MI) is a leading cause of morbidity worldwide, resulting from ischemic damage and necrosis to cardiomyocytes. While the standard treatment regimen for MI can be successful in restoring coronary perfusion, it typically does not resolve myocardial damage, which can leave patients particularly vulnerable to complications such as heart failure or electrical conduction abnormalities. Stem cell therapies offer a promising novel approach aimed at restoring cardiac function and decreasing the incidence of functional complications after an MI. This review used a literature search to evaluate the current landscape of stem cell therapy for post-MI recovery and focuses on the stem cell candidates for MI recovery therapy, delivery methods of such treatment, and their effectiveness. Both preclinical and clinical trials have demonstrated the safety of stem cells, but have struggled with limited cell retention, inconsistent efficacy, and survival. Mechanisms are employed by stem cells to promote regeneration, such as paracrine signaling, angiogenesis, and structural remodeling, in addition to the various stem cell delivery methods, including intracoronary infusion, direct myocardial injection, and intravenous administration. Furthermore, some strategies to combat past challenges in this field are discussed; for instance, extracellular vesicles, bioengineered patches, hydrogels, gene editing, and bioprinting. This article will provide a framework for future research in stem cell therapies and highlight the current progress in the field. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

14 pages, 6033 KiB  
Article
Ivosidenib Confers BRCAness Phenotype and Synthetic Lethality to Poly (ADP-Ribose) Polymerase Inhibition in BRCA1/2-Proficient Cancer Cells
by Danyang Zhou, Wei Liu, Yanyan Zhang and Chong Li
Biomedicines 2025, 13(4), 958; https://doi.org/10.3390/biomedicines13040958 - 14 Apr 2025
Viewed by 808
Abstract
Background/Objectives: PARP inhibitors (PARPi) are pivotal to treating homologous recombination repair-deficient (HRD) cancers, particularly BRCA1/2-mutated ovarian and breast cancers. However, most ovarian and breast cancers harbor wild-type (WT) BRCA1/2, limiting PARPi eligibility. This study aims to identify an approved drug [...] Read more.
Background/Objectives: PARP inhibitors (PARPi) are pivotal to treating homologous recombination repair-deficient (HRD) cancers, particularly BRCA1/2-mutated ovarian and breast cancers. However, most ovarian and breast cancers harbor wild-type (WT) BRCA1/2, limiting PARPi eligibility. This study aims to identify an approved drug that could induce a BRCAness phenotype, thereby sensitizing WT BRCA cancers to PARPi. Methods: Ovarian and breast cancer cell lines with WT BRCA1/2 were treated with ivosidenib. HR repair efficiency was assessed via RAD51 foci formation and reporter assays. Synthetic lethality with PARPi was evaluated using viability and colony formation assays. Mechanistic studies included RNA-binding protein pulldown, co-immunoprecipitation, and functional analyses of DNA repair pathways. YTHDC2′s role in HR was investigated through siRNA knockdown and rescue experiments. Results: Ivosidenib significantly reduced HR repair efficiency and sensitized cells to PARPi, inducing synthetic lethality. Mechanistically, ivosidenib directly bound YTHDC2, an m6A reader critical for HR. This interaction disrupted YTHDC2′s ability to promote DNA double-strand break repair via HR, evidenced by impaired recruitment of repair proteins (e.g., BRCA1, RAD51) and accumulation of DNA damage (γH2AX foci). YTHDC2 knockdown phenocopied ivosidenib effects, while overexpression rescued HR defects. Conclusions: Ivosidenib induces BRCAness in WT BRCA ovarian and breast cancers by targeting YTHDC2, thereby suppressing HR repair and enhancing PARPi sensitivity. This uncovers a novel, metabolism-independent mechanism of ivosidenib, repositioning it as a therapeutic agent for HRD tumors. These findings propose a strategy to expand PARPi eligibility to WT BRCA cancers, addressing a critical unmet need in oncology. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

20 pages, 9915 KiB  
Article
The Outcome of Cell Therapy Treating Urinary Incontinence Correlates with Precise Cell Localization in the Sphincter Complex
by Niklas Harland, Liv Johnen, Kamal T. Avula, Andrea Buzanich-Ladinig, Lukas Schwarz, Jasmin Knoll, Arnulf Stenzl and Wilhelm K. Aicher
Biomedicines 2025, 13(4), 917; https://doi.org/10.3390/biomedicines13040917 - 9 Apr 2025
Viewed by 511
Abstract
Background/Objectives: Urethral sphincter muscle deficiency is the leading cause of stress urinary incontinence. Preclinical and clinical studies suggested that cell therapy may improve the situation. However, the overall efficacy of cell therapies did often not satisfy the patient’s needs. We, therefore, investigated [...] Read more.
Background/Objectives: Urethral sphincter muscle deficiency is the leading cause of stress urinary incontinence. Preclinical and clinical studies suggested that cell therapy may improve the situation. However, the overall efficacy of cell therapies did often not satisfy the patient’s needs. We, therefore, investigated in a large animal model of incontinence if the localization of injected regenerative cells in the deficient urethral sphincter muscle correlated with the outcome. Methods: Urethral sphincter insufficiency was induced in three cohorts of pigs and confirmed by urodynamics. Then, either myogenic progenitor cells (MPCs) or adipose tissue-derived stromal cells (ADSCs) were injected into the injured sphincter complex by Williams needle under visual using a cystoscope. Sham-treated animals served as controls. Functional sphincter muscle regeneration was monitored by urodynamics over 5 weeks of follow-up. The localization of the injected cells was investigated by histology of cryosections of the tissue targeted. Results: Injection of MPCs near the sphincter muscle yielded better functional recovery when compared to MPC injections in adjacent sides. By contrast, injection of ADSCs in the submucosal tissue adjacent to the muscle led to better regeneration when compared to ADSC injections into the sphincter muscle. After five weeks of follow-up, MPCs yielded an overall robust but not significant improvement when compared to mock-treated controls, while ADSC injections reached significance. Conclusions: This small proof-of-principle study suggests that the clinical outcome of cell therapy for urinary incontinence depends on the choice of therapeutic cells and the precise localization of the cells in the tissue targeted as well. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

13 pages, 5358 KiB  
Article
Intra-Bone Marrow Administration of miR-140-3p Improves Bone Metabolism in a Growing Senescence-Accelerated Mouse Prone 6 Strain
by Hironobu Katsuyama, Kazue Tanda, Kumiko Terada, Shigeko Fushimi, Takeshi Yoda, Vitalii Katsuyama and Tsutomu Nohno
Biomedicines 2025, 13(4), 883; https://doi.org/10.3390/biomedicines13040883 - 5 Apr 2025
Viewed by 593
Abstract
Background: Our previous study demonstrated that miR-140-3p induced osteocalcin expression in osteoblastic MC3T3-E1 cells. In this study, we investigated the direct effects of miR-140-3p on bone turnover in senescence-accelerated mice. Methods: In order to evaluate the effects of miR-140-3p, we formulated [...] Read more.
Background: Our previous study demonstrated that miR-140-3p induced osteocalcin expression in osteoblastic MC3T3-E1 cells. In this study, we investigated the direct effects of miR-140-3p on bone turnover in senescence-accelerated mice. Methods: In order to evaluate the effects of miR-140-3p, we formulated lipid nanoparticles (LNPs) containing miR-140-3p (100 μg/mL), with or without flotillin-2 (Flo2), a microvesicle marker excreted by osteoblasts. LNP was administered into the right tibia of the P6 strain of senescence-accelerated mice (SAMP6). Four-week-old SAMP6 males were divided into three groups: control, LNP, and LNP + Flo2. LNPs were administered five times, once every three days. No gait abnormalities were observed in any group. Two days after the last administration of LNPs, blood and urine samples were collected to measure bone turnover markers and blood chemistry and to perform urinalysis. Bone histomorphometry was performed on the left femur, contralateral to the administration site. The pancreas was removed for insulin staining of the Langerhans islets. Results: The LNP + Flo2 group showed greater bone volume, trabecular thickness, and osteoid thickness in bone histomorphometry. Carboxylated osteocalcin, a bone formation marker, was also higher in the LNP + Flo2 group, indicating that LNP + Flo2 activated osteoblastic function. Insulin levels in the islets of Langerhans did not differ across the groups, consistent with under-carboxylated osteocalcin levels. Conclusions: LNP + Flo2 effectively improved bone metabolism. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

Back to TopTop