Objective: Fibronectin 1 (FN1) encodes fibronectin, a protein essential for cell adhesion, migration, extracellular matrix assembly, and regulation of cell differentiation and proliferation. While FN1 has been implicated in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), its role in neural differentiation remains unclear. This study aimed to investigate the effect of FN1 gene interference on neural differentiation of human BMSCs and explore the underlying molecular mechanisms.
Method: Three small interfering RNAs (ssi-417, si-4467 and si-5468) targeting FN1 were designed and transfected into BMSCs undergoing neural differentiation. Morphological changes were observed, and FN1 expression was assessed at both mRNA and protein levels. Alkaline phosphatase (ALP) staining was performed, and the expression of neural differentiation-related markers (MAP2, Tuj1, NSE and DCX) was quantified. Transcriptome sequencing was used to identify differentially expressed genes (DEGs), alternative splicing (AS) events and key pathways. Protein-protein interaction (PPI) network analysis was conducted to identify hub genes.
Result: Cells in the FN1 interference group retained a spindle-shaped mesenchymal morphology. FN1 expression at both mRNA and protein levels was significantly reduced in all three siRNA groups compared with the model group (P < 0.05). ALP staining showed a higher positive rate in the FN1 interference group. Expression of neural differentiation markers (MAP2, Tuj1, NSE and DCX) was significantly downregulated in the interference group compared with the model group (P < 0.05). Transcriptome analysis revealed 1047 upregulated and 1077 downregulated DEGs, enriched in pathways related to signal transduction, immune response, RNA processing, apoptosis and DNA repair. Additionally, 2246 alternative splicing events were identified, and PPI network analysis highlighted IL-6 as a core gene.
Conclusion: FN1 gene interference inhibits neural differentiation of BMSCs and alters key signaling pathways and splicing patterns, suggesting that FN1 plays a critical role in regulating stem cell fate. These findings provide new insights into the molecular mechanisms underlying neural differentiation of BMSCs.
{"title":"The role of FN1 gene interference in neural differentiation of human bone marrow mesenchymal stem cells.","authors":"Buhe Buren, Chaoqian Han, Chenyuan Yang, Feng Li, Dongsheng Fan, Xiaolong Wang, Xiaosai Hou, Xiaoxu Liu, Shangfei Jing","doi":"10.62347/TCNJ7008","DOIUrl":"10.62347/TCNJ7008","url":null,"abstract":"<p><strong>Objective: </strong>Fibronectin 1 (FN1) encodes fibronectin, a protein essential for cell adhesion, migration, extracellular matrix assembly, and regulation of cell differentiation and proliferation. While FN1 has been implicated in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), its role in neural differentiation remains unclear. This study aimed to investigate the effect of FN1 gene interference on neural differentiation of human BMSCs and explore the underlying molecular mechanisms.</p><p><strong>Method: </strong>Three small interfering RNAs (ssi-417, si-4467 and si-5468) targeting FN1 were designed and transfected into BMSCs undergoing neural differentiation. Morphological changes were observed, and FN1 expression was assessed at both mRNA and protein levels. Alkaline phosphatase (ALP) staining was performed, and the expression of neural differentiation-related markers (MAP2, Tuj1, NSE and DCX) was quantified. Transcriptome sequencing was used to identify differentially expressed genes (DEGs), alternative splicing (AS) events and key pathways. Protein-protein interaction (PPI) network analysis was conducted to identify hub genes.</p><p><strong>Result: </strong>Cells in the FN1 interference group retained a spindle-shaped mesenchymal morphology. FN1 expression at both mRNA and protein levels was significantly reduced in all three siRNA groups compared with the model group (<i>P</i> < 0.05). ALP staining showed a higher positive rate in the FN1 interference group. Expression of neural differentiation markers (MAP2, Tuj1, NSE and DCX) was significantly downregulated in the interference group compared with the model group (<i>P</i> < 0.05). Transcriptome analysis revealed 1047 upregulated and 1077 downregulated DEGs, enriched in pathways related to signal transduction, immune response, RNA processing, apoptosis and DNA repair. Additionally, 2246 alternative splicing events were identified, and PPI network analysis highlighted IL-6 as a core gene.</p><p><strong>Conclusion: </strong>FN1 gene interference inhibits neural differentiation of BMSCs and alters key signaling pathways and splicing patterns, suggesting that FN1 plays a critical role in regulating stem cell fate. These findings provide new insights into the molecular mechanisms underlying neural differentiation of BMSCs.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"201-216"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629971/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585686","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Type 2 diabetes mellitus (T2DM) is characterized by insulin resistance and β-cell dysfunction, with chronic inflammation playing a central pathogenic role. Mesenchymal stem cells (MSCs) possess therapeutic potential through immunomodulatory and tissue-reparative properties. This study aimed to evaluate the safety and efficacy of intravenous allogeneic umbilical cord-derived MSCs (UC-MSCs) in patients with T2DM.
Methods: Eleven adults with T2DM (disease duration ≥ 10 years; HbA1c ≤ 8%) received a single intravenous infusion of 1 × 108 UC-MSCs. This open-label pilot trial assessed safety (adverse events, hematologic and metabolic parameters) and efficacy (glycemic control and inflammatory gene expression) over a 2-month follow-up period. UC-MSCs were isolated under standardized conditions.
Results: UC-MSC transplantation in patients with T2DM was well tolerated, with only transient fever (36.3%) and mild muscle pain (18.2%) reported. The intervention resulted in significant metabolic improvements, including a 2.1% reduction in HbA1c (P = 0.00095) and a decrease in fasting glucose by 93.7 mg/dL (P = 0.00097). Treatment also modulated inflammatory pathways, as evidenced by upregulating of IKBα (1.76-fold, P = 0.0067) and downregulating of TNFα (0.62-fold) and IL-6 (0.65-fold). Variability in IKBα expression accounted for 48% of the variance in HbA1c (r = -0.69). Two distinct response patterns were observed: improvement in insulin sensitivity (7/11) via NF-κB suppression, and enhancement of β-cell function (3/11).
Conclusion: Allogeneic UC-MSC transplantation appears safe and significantly improves glycemic control in patients with T2DM. The heterogeneity in patient responses underscores the importance of stratification based on inflammatory status. These findings support UC-MSC therapy as a promising disease-modifying strategy and highlight the need for larger, controlled clinical trials.
{"title":"Safety and efficacy of allogeneic umbilical cord-derived mesenchymal stem cell transplantation in type 2 diabetes: a pilot clinical trial.","authors":"Ramin Raoufinia, Jalil Tavakol-Afshari, Mozhgan Afkhamizadeh, Ehsan Saburi, Amir Adhami Moghadam, Sareh Etemad, Hamid Reza Rahimi","doi":"10.62347/OPHF7871","DOIUrl":"10.62347/OPHF7871","url":null,"abstract":"<p><strong>Background: </strong>Type 2 diabetes mellitus (T2DM) is characterized by insulin resistance and β-cell dysfunction, with chronic inflammation playing a central pathogenic role. Mesenchymal stem cells (MSCs) possess therapeutic potential through immunomodulatory and tissue-reparative properties. This study aimed to evaluate the safety and efficacy of intravenous allogeneic umbilical cord-derived MSCs (UC-MSCs) in patients with T2DM.</p><p><strong>Methods: </strong>Eleven adults with T2DM (disease duration ≥ 10 years; HbA1c ≤ 8%) received a single intravenous infusion of 1 × 10<sup>8</sup> UC-MSCs. This open-label pilot trial assessed safety (adverse events, hematologic and metabolic parameters) and efficacy (glycemic control and inflammatory gene expression) over a 2-month follow-up period. UC-MSCs were isolated under standardized conditions.</p><p><strong>Results: </strong>UC-MSC transplantation in patients with T2DM was well tolerated, with only transient fever (36.3%) and mild muscle pain (18.2%) reported. The intervention resulted in significant metabolic improvements, including a 2.1% reduction in HbA1c (P = 0.00095) and a decrease in fasting glucose by 93.7 mg/dL (P = 0.00097). Treatment also modulated inflammatory pathways, as evidenced by upregulating of IKBα (1.76-fold, P = 0.0067) and downregulating of TNFα (0.62-fold) and IL-6 (0.65-fold). Variability in IKBα expression accounted for 48% of the variance in HbA1c (r = -0.69). Two distinct response patterns were observed: improvement in insulin sensitivity (7/11) via NF-κB suppression, and enhancement of β-cell function (3/11).</p><p><strong>Conclusion: </strong>Allogeneic UC-MSC transplantation appears safe and significantly improves glycemic control in patients with T2DM. The heterogeneity in patient responses underscores the importance of stratification based on inflammatory status. These findings support UC-MSC therapy as a promising disease-modifying strategy and highlight the need for larger, controlled clinical trials.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"244-260"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629970/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-15eCollection Date: 2025-01-01DOI: 10.62347/XFHZ3412
Zahid Habib Qureshi, Tahir Maqbool, Bakhtawar Farooq, Awais Altaf, Muzammal Mateen Azhar, Muhammad Rafiq, Muhammad Sarwar
Background: Liver diseases remain a major global health burden, with limited treatment options for advanced hepatic dysfunction. Stem cell-based therapies offer a favorable strategy for liver regeneration by providing a renewable source of functional hepatocyte-like cells (HLCs). This study aims to investigate the effect of Fibroblast growth factor (FGF) and Insulin-like Growth Factor (IGF) pre-treatment on the differentiation capacity of Umbilical Cord-Derived Mesenchymal Stem Cells (UC-MSCs) and their potential application in regenerative therapy for liver fibrosis or cirrhosis.
Methods: Cell viability was evaluated through MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), crystal violet, and trypan blue assays. For the assessment of differentiation potential, ELISA (Enzyme-Linked Immunosorbent Assay) and Immunocytochemistry of Hepatocyte Growth Factor (HGF) and Epidermal Growth Factor (EGF) were performed. For angiogenesis, an ELISA of Vascular Endothelial Growth Factor (VEGF) was performed. For apoptosis, an ELISA of p53 was performed. Gene expression analysis of differentiation markers, including Cytochrome P450 Family 1 Subfamily A Member 2 (CYP1A2), Cytochrome P450 Family 3 Subfamily A Member 2 (CYP3A2), Hepatocyte Growth Factor (HGF), Epidermal Growth Factor (EGF), Alkaline Phosphatase (ALP), Alpha-Fetoprotein (AFP), and albumin, was also performed. Furthermore, antioxidant enzymes were also measured.
Results: UC-MSCs preconditioned with FGF and IGF exhibited significantly enhanced viability and reduced cell death, as confirmed by MTT, crystal violet, and trypan blue assays. ELISA and immunocytochemistry demonstrated marked upregulation of hepatic markers (HGF, EGF), angiogenic factor (VEGF), and reduced expression of the apoptotic marker p53 in the preconditioned groups. The gene expression analysis confirmed superior regenerative potential in the FGF+IGF-treated group. Antioxidative analysis further validated a higher level of antioxidative potential in preconditioned cells.
Conclusion: Preconditioned UC-MSCs offer a promising cell-based alternative to liver transplantation by enhancing regeneration, reducing apoptosis, and promoting angiogenesis and antioxidant defense in damaged liver tissue.
{"title":"Synergistic growth factors preconditioning strategy to improve hepatic differentiation efficiency of UC-MSCs <i>in vitro</i>.","authors":"Zahid Habib Qureshi, Tahir Maqbool, Bakhtawar Farooq, Awais Altaf, Muzammal Mateen Azhar, Muhammad Rafiq, Muhammad Sarwar","doi":"10.62347/XFHZ3412","DOIUrl":"10.62347/XFHZ3412","url":null,"abstract":"<p><strong>Background: </strong>Liver diseases remain a major global health burden, with limited treatment options for advanced hepatic dysfunction. Stem cell-based therapies offer a favorable strategy for liver regeneration by providing a renewable source of functional hepatocyte-like cells (HLCs). This study aims to investigate the effect of Fibroblast growth factor (FGF) and Insulin-like Growth Factor (IGF) pre-treatment on the differentiation capacity of Umbilical Cord-Derived Mesenchymal Stem Cells (UC-MSCs) and their potential application in regenerative therapy for liver fibrosis or cirrhosis.</p><p><strong>Methods: </strong>Cell viability was evaluated through MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), crystal violet, and trypan blue assays. For the assessment of differentiation potential, ELISA (Enzyme-Linked Immunosorbent Assay) and Immunocytochemistry of Hepatocyte Growth Factor (HGF) and Epidermal Growth Factor (EGF) were performed. For angiogenesis, an ELISA of Vascular Endothelial Growth Factor (VEGF) was performed. For apoptosis, an ELISA of p53 was performed. Gene expression analysis of differentiation markers, including Cytochrome P450 Family 1 Subfamily A Member 2 (CYP1A2), Cytochrome P450 Family 3 Subfamily A Member 2 (CYP3A2), Hepatocyte Growth Factor (HGF), Epidermal Growth Factor (EGF), Alkaline Phosphatase (ALP), Alpha-Fetoprotein (AFP), and albumin, was also performed. Furthermore, antioxidant enzymes were also measured.</p><p><strong>Results: </strong>UC-MSCs preconditioned with FGF and IGF exhibited significantly enhanced viability and reduced cell death, as confirmed by MTT, crystal violet, and trypan blue assays. ELISA and immunocytochemistry demonstrated marked upregulation of hepatic markers (HGF, EGF), angiogenic factor (VEGF), and reduced expression of the apoptotic marker p53 in the preconditioned groups. The gene expression analysis confirmed superior regenerative potential in the FGF+IGF-treated group. Antioxidative analysis further validated a higher level of antioxidative potential in preconditioned cells.</p><p><strong>Conclusion: </strong>Preconditioned UC-MSCs offer a promising cell-based alternative to liver transplantation by enhancing regeneration, reducing apoptosis, and promoting angiogenesis and antioxidant defense in damaged liver tissue.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"187-200"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629968/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585719","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-15eCollection Date: 2025-01-01DOI: 10.62347/RQXG2881
Amoura M Abou-El-Naga, Mohamed A M Sobh, Mohamed M Fathy, Ahmed M Badawy, Ahmed Hk El-Hashash
Background: The ovarian surface epithelium (OSE) stem cells are crucial components of the human ovary and play a significant role in both the reproductive function and ovulatory wound repair. Harnessing these stem cells could provide a novel therapeutic strategy for reproductive disorders.
Methods: In this study, we determine and compare the differentiation potential of OSE stem cells into Oocyte-like cells between human and mice-derived OSE stem cells. In addition, we assessed OSE cellular characteristics associated with their stemness and self-renewal abilities and demonstrated their capacity for in vitro differentiation.
Results: We found that the ovarian surface epithelium harbored putative stem cells characterized by Alkaline Phosphatase (AP) activity, cell proliferation, expression of mesenchymal lineage surface markers, and pluripotent transcriptional markers. Interestingly, human-derived OSE stem cells exhibited increased AP activity and cell proliferation compared to mouse OSE stem cells, suggesting high levels of self-renewal and differentiation potential. Moreover, our evaluation of the in vitro differentiation potential into Oocyte-like cells for human or mouse OSE stem cells demonstrated an enhanced oogenesis potential for human OSE stem cells compared to mouse OSE cells, as evidenced by the analysis of germ cell marker expression and the production of Oocyte-like cells.
Conclusion: Our data highlighted the difference in the characterization and differentiation potential into Oocyte-like cells between human-derived OSE stem cells and mouse-derived OSE stem cells and lay a foundation for a future establishment of stem cell line with implications for reproductive cell therapy. In the realm of reproductive medicine, infertile patients with nonfunctional ovaries represent a significant area of interest, and any potential to regenerate their ovaries would hold great importance.
{"title":"Comparison of differentiation potential of ovarian surface epithelial stem cells into Oocyte-like cells between human- and mice-derived cells.","authors":"Amoura M Abou-El-Naga, Mohamed A M Sobh, Mohamed M Fathy, Ahmed M Badawy, Ahmed Hk El-Hashash","doi":"10.62347/RQXG2881","DOIUrl":"10.62347/RQXG2881","url":null,"abstract":"<p><strong>Background: </strong>The ovarian surface epithelium (OSE) stem cells are crucial components of the human ovary and play a significant role in both the reproductive function and ovulatory wound repair. Harnessing these stem cells could provide a novel therapeutic strategy for reproductive disorders.</p><p><strong>Methods: </strong>In this study, we determine and compare the differentiation potential of OSE stem cells into Oocyte-like cells between human and mice-derived OSE stem cells. In addition, we assessed OSE cellular characteristics associated with their stemness and self-renewal abilities and demonstrated their capacity for in vitro differentiation.</p><p><strong>Results: </strong>We found that the ovarian surface epithelium harbored putative stem cells characterized by Alkaline Phosphatase (AP) activity, cell proliferation, expression of mesenchymal lineage surface markers, and pluripotent transcriptional markers. Interestingly, human-derived OSE stem cells exhibited increased AP activity and cell proliferation compared to mouse OSE stem cells, suggesting high levels of self-renewal and differentiation potential. Moreover, our evaluation of the in vitro differentiation potential into Oocyte-like cells for human or mouse OSE stem cells demonstrated an enhanced oogenesis potential for human OSE stem cells compared to mouse OSE cells, as evidenced by the analysis of germ cell marker expression and the production of Oocyte-like cells.</p><p><strong>Conclusion: </strong>Our data highlighted the difference in the characterization and differentiation potential into Oocyte-like cells between human-derived OSE stem cells and mouse-derived OSE stem cells and lay a foundation for a future establishment of stem cell line with implications for reproductive cell therapy. In the realm of reproductive medicine, infertile patients with nonfunctional ovaries represent a significant area of interest, and any potential to regenerate their ovaries would hold great importance.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"170-186"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585896","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-15eCollection Date: 2025-01-01DOI: 10.62347/GZVM7194
Nicholas Panin, Luis Torres, Yug Patel, Alana Michelson, Amanda Hung, Ahmed H El-Hashash
Stem cell therapy is revolutionizing the treatment of neurological disorders, offering innovative approaches for regeneration and repair. This paper explores five distinct mechanisms of stem cell therapy, focusing on their applications and therapeutic potential. Neural stem cells (NSCs) combined with pharmacological agents, such as FTY720, enhance remyelination and neural repair in multiple sclerosis (MS) and spinal cord injuries (SCI). Induced pluripotent stem cells (iPSCs) provide a personalized approach by enabling the generation of patient-specific NSCs for treating conditions like Parkinson's Disease (PD). Gene-editing technologies, such as CRISPR-Cas9, expand the scope of NSC applications by facilitating precise interventions for genetic disorders like SMARD1. Neurotrophic factors derived from NSCs present a cell-free alternative to promote neuronal survival and repair in diseases such as Parkinson's and Huntington's disease. Additionally, NSC-derived extracellular vesicle therapies, such as intranasal delivery methods for AD treatment, offer non-invasive approaches to reduce neuroinflammation and enhance cognitive recovery. While these mechanisms demonstrate remarkable therapeutic potential, challenges such as cost, scalability, and safety remain. This review provides a comprehensive analysis of these mechanisms, highlighting their contributions to the future of regenerative medicine and personalized therapeutic strategies.
{"title":"Innovative approaches in neural stem cell therapy: a comprehensive review of mechanisms and applications.","authors":"Nicholas Panin, Luis Torres, Yug Patel, Alana Michelson, Amanda Hung, Ahmed H El-Hashash","doi":"10.62347/GZVM7194","DOIUrl":"10.62347/GZVM7194","url":null,"abstract":"<p><p>Stem cell therapy is revolutionizing the treatment of neurological disorders, offering innovative approaches for regeneration and repair. This paper explores five distinct mechanisms of stem cell therapy, focusing on their applications and therapeutic potential. Neural stem cells (NSCs) combined with pharmacological agents, such as FTY720, enhance remyelination and neural repair in multiple sclerosis (MS) and spinal cord injuries (SCI). Induced pluripotent stem cells (iPSCs) provide a personalized approach by enabling the generation of patient-specific NSCs for treating conditions like Parkinson's Disease (PD). Gene-editing technologies, such as CRISPR-Cas9, expand the scope of NSC applications by facilitating precise interventions for genetic disorders like SMARD1. Neurotrophic factors derived from NSCs present a cell-free alternative to promote neuronal survival and repair in diseases such as Parkinson's and Huntington's disease. Additionally, NSC-derived extracellular vesicle therapies, such as intranasal delivery methods for AD treatment, offer non-invasive approaches to reduce neuroinflammation and enhance cognitive recovery. While these mechanisms demonstrate remarkable therapeutic potential, challenges such as cost, scalability, and safety remain. This review provides a comprehensive analysis of these mechanisms, highlighting their contributions to the future of regenerative medicine and personalized therapeutic strategies.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"230-243"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629974/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585646","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-15eCollection Date: 2025-01-01DOI: 10.62347/FTXA8845
Chadwick C Prodromos, Ruby Del Villar, Max Y Jin, Alaa Abd-Elsayed, Kenneth Candido
Aim: Amyotrophic lateral sclerosis (ALS), Lewy Body dementia (LBD), Kennedy disease (KD), and Congenital Myasthenic Syndrome (CMS) are progressive motor disorders for which no disease modifying treatment exists. ALS and LBD are uniformly, and often rapidly, fatal. No treatment of any kind has ever resulted in actual improvement for ALS patients; the best that has been achieved is minor slowing of their progression. Forty-one preclinical studies of intra-nasal instillation of mesenchymal stem cell exosomes have, however, demonstrated complete safety and efficacy for models of a variety of neurocognitive and motor disorders. We hypothesized that intranasal exosomes treatment in humans would be completely safe and also effective for the treatment of motor disorders such as ALS, LBD, KD and CMS.
Methods: 18 patients with ALS, Kennedy Disease, Congenital Myasthenic Syndrome, or Lewy Body Dementia had 32 AlloEx Exosome® treatments to assess safety, attenuation of disease, and increase in strength and motor function. The study was conducted under the clinical trial NCT07105371 found at clinicaltrials.gov/study/NCT07105371.
Results: There were no adverse events of any kind reported among these treatments. All patients, except for one, achieved some degree of clinical and strength improvement; the longest improvement was recorded at the 6-month follow-up.
Conclusion: Intranasally-instilled AlloEx Exosomes® are completely safe, attenuate progression, and improve strength in ALS, Kennedy Disease, CMS, and LBD.
{"title":"Exosome-rich mesenchymal stem cell secretome improves strength in patients with amyotrophic lateral sclerosis, Kennedy disease, congenital myasthenic syndrome and Lewy body dementia.","authors":"Chadwick C Prodromos, Ruby Del Villar, Max Y Jin, Alaa Abd-Elsayed, Kenneth Candido","doi":"10.62347/FTXA8845","DOIUrl":"10.62347/FTXA8845","url":null,"abstract":"<p><strong>Aim: </strong>Amyotrophic lateral sclerosis (ALS), Lewy Body dementia (LBD), Kennedy disease (KD), and Congenital Myasthenic Syndrome (CMS) are progressive motor disorders for which no disease modifying treatment exists. ALS and LBD are uniformly, and often rapidly, fatal. No treatment of any kind has ever resulted in actual improvement for ALS patients; the best that has been achieved is minor slowing of their progression. Forty-one preclinical studies of intra-nasal instillation of mesenchymal stem cell exosomes have, however, demonstrated complete safety and efficacy for models of a variety of neurocognitive and motor disorders. We hypothesized that intranasal exosomes treatment in humans would be completely safe and also effective for the treatment of motor disorders such as ALS, LBD, KD and CMS.</p><p><strong>Methods: </strong>18 patients with ALS, Kennedy Disease, Congenital Myasthenic Syndrome, or Lewy Body Dementia had 32 AlloEx Exosome<sup>®</sup> treatments to assess safety, attenuation of disease, and increase in strength and motor function. The study was conducted under the clinical trial NCT07105371 found at clinicaltrials.gov/study/NCT07105371.</p><p><strong>Results: </strong>There were no adverse events of any kind reported among these treatments. All patients, except for one, achieved some degree of clinical and strength improvement; the longest improvement was recorded at the 6-month follow-up.</p><p><strong>Conclusion: </strong>Intranasally-instilled AlloEx Exosomes<sup>®</sup> are completely safe, attenuate progression, and improve strength in ALS, Kennedy Disease, CMS, and LBD.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"217-229"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-15eCollection Date: 2025-01-01DOI: 10.62347/AMKH1816
Haojiang Li, Jianfei Gao, Ren Zhang, Jie Liu, Haiquan Tian, Yujia Xin, Xiaoliang Song, Xiangyi Li, Yuewen He
Objective: The aim of this study was to identify the different immune-related genes (DIRGs) of mesenchymal stem cells (MSCs) in three-dimensional (3D) vs. two-dimensional (3D) environment.
Materials and methods: The gene expression dataset GSE52896 was downloaded from the Gene Expression Omnibus (GEO) database. We obtained immune-related genes from the ImmPort database. The array was processed with the R language to obtain differentially expressed genes (DEGs). A protein-protein interaction (PPI) network was constructed with the STRING database and analyzed with Cytoscape. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis data were performed with DAVID (https://davidbioinformatics.nih.gov/). We constructed a least absolute shrinkage and selection operator (LASSO) regression model and multiple support vector machine - recursive feature elimination (mSVM-RFE) model to identify the key DIRGs in cells growing in 3D culture. The performance of the key genes was validated in the GSE58919 dataset. Western blot analysis was performed to verify the expression of one key gene, Cysteine and Glycine Rich Protein 1 (CSRP1). Key immune-related genes were identified using CIBERSORT (https://cibersortx.stanford.edu/).
Results: A total of 446 DEGs were screened under two different culture conditions (2D and 3D), and 65 DEGs were identified. GO analysis revealed changes in inflammatory response, extracellular region, and protein binding. KEGG enrichment analysis showed that the DEGs were enriched in pathways involved in cytokine-cytokine receptor interactions, viral protein interactions with cytokines and cytokine receptors and the TNF signaling pathway. Seven key genes were obtained from the intersection of the outputs of the LASSO and mSVM-RFE algorithms. The expression of the seven key genes was verified in the GSE52896 dataset. Western blot (WB) confirmed the alteration of CSRP1 expression under different culture conditions.
Conclusion: Stem cells showed significant changes in immune response gene expression under 3D culture conditions. CSRP1 plays essential roles in MSC immunomodulation.
目的:本研究的目的是鉴定三维(3D)和二维(3D)环境下间充质干细胞(MSCs)不同的免疫相关基因(DIRGs)。材料和方法:从gene expression Omnibus (GEO)数据库下载基因表达数据集GSE52896。我们从import数据库中获得免疫相关基因。用R语言对该阵列进行处理,获得差异表达基因(DEGs)。利用STRING数据库构建蛋白-蛋白相互作用(PPI)网络,并用Cytoscape软件进行分析。基因本体(GO)和京都基因与基因组百科全书(KEGG)途径富集分析数据使用DAVID (https://davidbioinformatics.nih.gov/)进行。我们构建了最小绝对收缩和选择算子(LASSO)回归模型和多支持向量机-递归特征消除(mSVM-RFE)模型来识别三维培养细胞中的关键dirg。在GSE58919数据集中验证了关键基因的性能。Western blot分析验证了一个关键基因——半胱氨酸和甘氨酸富蛋白1 (CSRP1)的表达。利用CIBERSORT (https://cibersortx.stanford.edu/).Results)对关键免疫相关基因进行鉴定:在2D和3D两种不同培养条件下共筛选446个基因,鉴定出65个基因。氧化石墨烯分析显示炎症反应、细胞外区域和蛋白质结合的变化。KEGG富集分析显示,在细胞因子-细胞因子受体相互作用、病毒蛋白与细胞因子和细胞因子受体相互作用以及TNF信号通路中,deg富集。从LASSO和mSVM-RFE算法的输出交集中获得7个关键基因。在GSE52896数据集中验证了7个关键基因的表达。Western blot (WB)证实了不同培养条件下CSRP1表达的改变。结论:干细胞在三维培养条件下免疫应答基因表达发生明显变化。CSRP1在MSC免疫调节中起重要作用。
{"title":"Analysis of key immune genes in mesenchymal stem cells in a 3D environment.","authors":"Haojiang Li, Jianfei Gao, Ren Zhang, Jie Liu, Haiquan Tian, Yujia Xin, Xiaoliang Song, Xiangyi Li, Yuewen He","doi":"10.62347/AMKH1816","DOIUrl":"10.62347/AMKH1816","url":null,"abstract":"<p><strong>Objective: </strong>The aim of this study was to identify the different immune-related genes (DIRGs) of mesenchymal stem cells (MSCs) in three-dimensional (3D) vs. two-dimensional (3D) environment.</p><p><strong>Materials and methods: </strong>The gene expression dataset GSE52896 was downloaded from the Gene Expression Omnibus (GEO) database. We obtained immune-related genes from the ImmPort database. The array was processed with the R language to obtain differentially expressed genes (DEGs). A protein-protein interaction (PPI) network was constructed with the STRING database and analyzed with Cytoscape. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis data were performed with DAVID (https://davidbioinformatics.nih.gov/). We constructed a least absolute shrinkage and selection operator (LASSO) regression model and multiple support vector machine - recursive feature elimination (mSVM-RFE) model to identify the key DIRGs in cells growing in 3D culture. The performance of the key genes was validated in the GSE58919 dataset. Western blot analysis was performed to verify the expression of one key gene, Cysteine and Glycine Rich Protein 1 (CSRP1). Key immune-related genes were identified using CIBERSORT (https://cibersortx.stanford.edu/).</p><p><strong>Results: </strong>A total of 446 DEGs were screened under two different culture conditions (2D and 3D), and 65 DEGs were identified. GO analysis revealed changes in inflammatory response, extracellular region, and protein binding. KEGG enrichment analysis showed that the DEGs were enriched in pathways involved in cytokine-cytokine receptor interactions, viral protein interactions with cytokines and cytokine receptors and the TNF signaling pathway. Seven key genes were obtained from the intersection of the outputs of the LASSO and mSVM-RFE algorithms. The expression of the seven key genes was verified in the GSE52896 dataset. Western blot (WB) confirmed the alteration of CSRP1 expression under different culture conditions.</p><p><strong>Conclusion: </strong>Stem cells showed significant changes in immune response gene expression under 3D culture conditions. CSRP1 plays essential roles in MSC immunomodulation.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 4","pages":"261-276"},"PeriodicalIF":1.9,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12629972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145585955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The association of bone marrow stem cells (BMSCs) with cardiac function outcomes and treatment outcomes in heart failure (HF) patients with low ejection fraction (EF) has been heterogeneous across studies. This systematic review aimed to investigate the effect of BMSCs on functional, clinical, quality of life, and major adverse cardiovascular events (MACE) outcomes in HF patients with low EF. PubMed, Scopus, Clinicaltrial.gov, Cochrane Library, Google Scholar, and Web and reference databases were searched for articles that examined the effect of BMSCs therapy on improving cardiac outcomes in patients with low EF, from 2000 to 2024. Differences in left ventricular ejection fraction (LVEF), MACE, echocardiographic indices (left ventricular ejection fraction (LVEF), left ventricular end-diastolic volume (LVEDV), and left ventricular end-systolic volume (LVESV)), 6-min walk test (6-MWT), New York Heart Association (NYHA) class and immunologic responses were defined as outcomes. Low EF was defined as an EF <45%. Finally, 14 RCTs involving 710 HF patients with low EF were included. BMSCs transplantation was associated with improvements in echocardiographic parameters, EF rate, and NYHA class in most studies (9 of 14) compared to the control group, regardless of the time of outcome assessment (3 or 6 months). It also significantly improved the 6-MWT in most studies. Improvements in parameters and functional outcomes were similar at both evaluation periods, 6 and 12 months. The BMSCs transplantation was not significantly associated with the incidence of MACE and immunological responses. The results of this systematic review supported the positive role of BMSCs transplantation in improving echocardiographic parameters, EF rate, NYHA class, and 6-MWT in HF patients with low EF. BMSCs transplantation was not significantly associated with the incidence of MACE and immunological responses.
{"title":"Bone marrow stem cell therapy in heart failure patients with low ejection fraction: a systematic review.","authors":"Azin Alizadehasl, Seyedeh Fatemeh Hosseini Jebelli, Yasamin Afsari Zonooz, Azam Yalameh Aliabadi, Maryam Mohseni Salehi, Golnar Mortaz Hejri, Sarah Forati, Hoda Hakimian","doi":"10.62347/YKPS8756","DOIUrl":"10.62347/YKPS8756","url":null,"abstract":"<p><p>The association of bone marrow stem cells (BMSCs) with cardiac function outcomes and treatment outcomes in heart failure (HF) patients with low ejection fraction (EF) has been heterogeneous across studies. This systematic review aimed to investigate the effect of BMSCs on functional, clinical, quality of life, and major adverse cardiovascular events (MACE) outcomes in HF patients with low EF. PubMed, Scopus, Clinicaltrial.gov, Cochrane Library, Google Scholar, and Web and reference databases were searched for articles that examined the effect of BMSCs therapy on improving cardiac outcomes in patients with low EF, from 2000 to 2024. Differences in left ventricular ejection fraction (LVEF), MACE, echocardiographic indices (left ventricular ejection fraction (LVEF), left ventricular end-diastolic volume (LVEDV), and left ventricular end-systolic volume (LVESV)), 6-min walk test (6-MWT), New York Heart Association (NYHA) class and immunologic responses were defined as outcomes. Low EF was defined as an EF <45%. Finally, 14 RCTs involving 710 HF patients with low EF were included. BMSCs transplantation was associated with improvements in echocardiographic parameters, EF rate, and NYHA class in most studies (9 of 14) compared to the control group, regardless of the time of outcome assessment (3 or 6 months). It also significantly improved the 6-MWT in most studies. Improvements in parameters and functional outcomes were similar at both evaluation periods, 6 and 12 months. The BMSCs transplantation was not significantly associated with the incidence of MACE and immunological responses. The results of this systematic review supported the positive role of BMSCs transplantation in improving echocardiographic parameters, EF rate, NYHA class, and 6-MWT in HF patients with low EF. BMSCs transplantation was not significantly associated with the incidence of MACE and immunological responses.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 3","pages":"146-155"},"PeriodicalIF":1.9,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12444438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145111881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Epigenetic modifiers play an important role in regulating the fate of hematopoietic stem cells (HSCs). The chromatin-modifying agents (CMA) have previously been shown to expand HSCs from cord blood (CB) and bone marrow (BM) CD34+ cells. Meanwhile, DNA methylation maintains persistent cellular memories and is thought to be the primary epigenetic barrier to reprogramming. The DNA hypomethylation drug decitabine is one of the CMA that could alter gene expression and HSC self-renewal. It has been reported that decitabine could promote platelets generation in ITP patients.
Objective: It's unknown if decitabine could affect CD34+ cells and megakaryocytes generation and maturation from human induced pluripotent stem cells (hiPSCs).
Methods: We utilized serum free, exon free and feeder free differentiation system to generate CD34+ from hiPSCs and induced them differentiation into megakaryocytes. Different concentrations of decitabine were added at different stages and analyzed these cells by RT-PCR, flow cytometry analysis, cell counting and other regular experimental methods.
Results: The proliferation and function of CD34+ cells in vitro were significantly suspended after exposure to decitabine. Low concentration of decitabine could maintain the CD34+ function. In addition, we found that decitabine did not have any effect on the megakaryocyte generation, but it prevented megakaryocyte maturation. The DNA methyltransferases (DNMTs) changed a lot not only in CD34+ stage but also in the megakaryocyte generation and maturation due to decitabine addition.
Conclusions: These results suggested that the effect of decitabine on CD34+ cells from hiPSCs was very different from CB, PB and BM CD34+ cells and the epigenetic changes may play an important role in the CD34+ expansion and megakaryocytes maturation. It may provide a potential mechanism of studying hiPSCs derived HSCs and megakaryocytes maturation in the future.
{"title":"The effect of decitabine on human induced pluripotent stem cells (hiPSCs) derived CD34<sup>+</sup> cells expansion and the megakaryocytes generation and maturation.","authors":"Yanfeng Liu, Hongyan Zhang, Yanxin Li, Chuanli Zhao","doi":"10.62347/IRBE1598","DOIUrl":"10.62347/IRBE1598","url":null,"abstract":"<p><strong>Background: </strong>Epigenetic modifiers play an important role in regulating the fate of hematopoietic stem cells (HSCs). The chromatin-modifying agents (CMA) have previously been shown to expand HSCs from cord blood (CB) and bone marrow (BM) CD34<sup>+</sup> cells. Meanwhile, DNA methylation maintains persistent cellular memories and is thought to be the primary epigenetic barrier to reprogramming. The DNA hypomethylation drug decitabine is one of the CMA that could alter gene expression and HSC self-renewal. It has been reported that decitabine could promote platelets generation in ITP patients.</p><p><strong>Objective: </strong>It's unknown if decitabine could affect CD34<sup>+</sup> cells and megakaryocytes generation and maturation from human induced pluripotent stem cells (hiPSCs).</p><p><strong>Methods: </strong>We utilized serum free, exon free and feeder free differentiation system to generate CD34<sup>+</sup> from hiPSCs and induced them differentiation into megakaryocytes. Different concentrations of decitabine were added at different stages and analyzed these cells by RT-PCR, flow cytometry analysis, cell counting and other regular experimental methods.</p><p><strong>Results: </strong>The proliferation and function of CD34<sup>+</sup> cells in vitro were significantly suspended after exposure to decitabine. Low concentration of decitabine could maintain the CD34<sup>+</sup> function. In addition, we found that decitabine did not have any effect on the megakaryocyte generation, but it prevented megakaryocyte maturation. The DNA methyltransferases (<i>DNMTs</i>) changed a lot not only in CD34<sup>+</sup> stage but also in the megakaryocyte generation and maturation due to decitabine addition.</p><p><strong>Conclusions: </strong>These results suggested that the effect of decitabine on CD34<sup>+</sup> cells from hiPSCs was very different from CB, PB and BM CD34<sup>+</sup> cells and the epigenetic changes may play an important role in the CD34<sup>+</sup> expansion and megakaryocytes maturation. It may provide a potential mechanism of studying hiPSCs derived HSCs and megakaryocytes maturation in the future.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 3","pages":"156-169"},"PeriodicalIF":1.9,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12444434/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145111906","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-25eCollection Date: 2025-01-01DOI: 10.62347/GQZH3508
Wenli Zhou, Xuehai Liu, Zhaoyu Li, Binkui Jia, Xilin Lei, Kai Sun, Pengfei Yang, Shiye He, Di Wang, Haoling Zhang, Sinong Wang
Stem cells possess self-renewal and multipotent differentiation capabilities, exhibiting broad applications in regenerative medicine and tissue homeostasis maintenance. Their fate regulation relies heavily on precise epigenetic mechanisms. Cancer stem cells (CSCs), as key drivers of tumor heterogeneity, recurrence, and drug resistance, share extensive epigenetic features with normal stem cells, forming a complex and dynamic regulatory network. Mechanisms including DNA methylation, histone modification, chromatin remodeling, and ncRNAs collectively sustain stem cell pluripotency and tumor stemness, while aberrant epigenetic alterations serve as core drivers of tumor initiation and progression. In recent years, with the advent of single-cell omics and CRISPR-dCas9 epigenetic editing technologies, epigenetic "crosstalk" between stem cells and tumor cells has been progressively uncovered, especially the multidimensional epigenetic reprogramming induced by the tumor microenvironment (TME) that promotes CSC traits and drug resistance. This review systematically summarizes the epigenetic regulatory mechanisms of stem cells, epigenetic abnormalities in tumors, their interactions, and translational potential in therapeutic strategies, focusing on frontier topics such as reversible epigenetic plasticity, metabolic-epigenetic interplay, and liquid biopsy epigenetic biomarkers. Looking forward, artificial intelligence (AI) and big data analysis are expected to deepen the understanding of epigenetic heterogeneity, driving integrative innovations in precision medicine and regenerative interventions. Comprehensive understanding of the epigenetic crosstalk between stem cells and tumors will provide solid theoretical support and technical pathways for CSC-targeted therapies, epigenetic drug development, and stem cell fate manipulation.
{"title":"Epigenetic crosstalk between stem cells and tumors: mechanisms and emerging perspectives.","authors":"Wenli Zhou, Xuehai Liu, Zhaoyu Li, Binkui Jia, Xilin Lei, Kai Sun, Pengfei Yang, Shiye He, Di Wang, Haoling Zhang, Sinong Wang","doi":"10.62347/GQZH3508","DOIUrl":"10.62347/GQZH3508","url":null,"abstract":"<p><p>Stem cells possess self-renewal and multipotent differentiation capabilities, exhibiting broad applications in regenerative medicine and tissue homeostasis maintenance. Their fate regulation relies heavily on precise epigenetic mechanisms. Cancer stem cells (CSCs), as key drivers of tumor heterogeneity, recurrence, and drug resistance, share extensive epigenetic features with normal stem cells, forming a complex and dynamic regulatory network. Mechanisms including DNA methylation, histone modification, chromatin remodeling, and ncRNAs collectively sustain stem cell pluripotency and tumor stemness, while aberrant epigenetic alterations serve as core drivers of tumor initiation and progression. In recent years, with the advent of single-cell omics and CRISPR-dCas9 epigenetic editing technologies, epigenetic \"crosstalk\" between stem cells and tumor cells has been progressively uncovered, especially the multidimensional epigenetic reprogramming induced by the tumor microenvironment (TME) that promotes CSC traits and drug resistance. This review systematically summarizes the epigenetic regulatory mechanisms of stem cells, epigenetic abnormalities in tumors, their interactions, and translational potential in therapeutic strategies, focusing on frontier topics such as reversible epigenetic plasticity, metabolic-epigenetic interplay, and liquid biopsy epigenetic biomarkers. Looking forward, artificial intelligence (AI) and big data analysis are expected to deepen the understanding of epigenetic heterogeneity, driving integrative innovations in precision medicine and regenerative interventions. Comprehensive understanding of the epigenetic crosstalk between stem cells and tumors will provide solid theoretical support and technical pathways for CSC-targeted therapies, epigenetic drug development, and stem cell fate manipulation.</p>","PeriodicalId":7657,"journal":{"name":"American journal of stem cells","volume":"14 3","pages":"98-133"},"PeriodicalIF":1.9,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12444435/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145111850","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}