首页 > 最新文献

Stem Cell Reviews and Reports最新文献

英文 中文
Human Umbilical Cord Mesenchymal Stem Cells for the Treatment of Systemic Lupus Erythematosus via Glucose Metabolism of CD4+T Cells.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-02-20 DOI: 10.1007/s12015-025-10848-1
Meng Ding, Lu Jin, Shaoxin Cui, Lin Yang, Jingjing He, Xiaoping Wang, Fei Chang, Qun Wang, Xue Liu, Hongtao Jin, Shuran Song, Min Shi, Jingjing Yu, Jun Ma, Aijing Liu

Background: T cells play a crucial role in the pathogenesis of systemic lupus erythematosus (SLE), with their functions regulated by various metabolic pathways. This study explores SLE pathogenesis and the therapeutic effects of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) via metabolic reprogramming.

Methods: Clinical data and peripheral blood samples were collected from 15 SLE patients and matched healthy controls. CD4+ T cells were isolated and activated in vitro with anti-CD3/CD28. Following 72 h of co-culture with hUC-MSCs, CD4+ T cell viability was assessed using the CCK-8 assay. The oxygen consumption rate (OCR) and glycolytic proton efflux rate (glycoPER) were measured with a Seahorse analyzer. Cytokine levels were detected by multiplex assay, and transcriptome sequencing was performed. Western blotting analyzed glucose metabolism-related enzymes and signaling pathways in lupus model mice.

Results: Compared to healthy controls, activated CD4+ T cells from SLE patients exhibited significantly increased OCR and glycoPER levels (P < 0.05). Following 72 h of co-culture with hUC-MSCs, OCR, glycoPER, cell viability, and pro-inflammatory factors in SLE-CD4+ T cells decreased markedly (P < 0.01). Upregulation of 434 genes and downregulation of 172 genes was observed, particularly in the JAK-STAT and PI3K-Akt pathways. hUC-MSCs inhibited the expression of glucose metabolism-related enzymes and the JAK-STAT and PI3K-Akt signaling pathways in lupus model mice.

Conclusion: hUC-MSCs inhibited the proliferation and function of aberrant CD4+ T cells in SLE patients by modulating glycometabolism and the JAK-STAT and PI3K-Akt signaling pathways, providing new insights into the therapeutic mechanisms of MSCs based on metabolic reprogramming.

{"title":"Human Umbilical Cord Mesenchymal Stem Cells for the Treatment of Systemic Lupus Erythematosus via Glucose Metabolism of CD4<sup>+</sup>T Cells.","authors":"Meng Ding, Lu Jin, Shaoxin Cui, Lin Yang, Jingjing He, Xiaoping Wang, Fei Chang, Qun Wang, Xue Liu, Hongtao Jin, Shuran Song, Min Shi, Jingjing Yu, Jun Ma, Aijing Liu","doi":"10.1007/s12015-025-10848-1","DOIUrl":"https://doi.org/10.1007/s12015-025-10848-1","url":null,"abstract":"<p><strong>Background: </strong>T cells play a crucial role in the pathogenesis of systemic lupus erythematosus (SLE), with their functions regulated by various metabolic pathways. This study explores SLE pathogenesis and the therapeutic effects of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) via metabolic reprogramming.</p><p><strong>Methods: </strong>Clinical data and peripheral blood samples were collected from 15 SLE patients and matched healthy controls. CD4<sup>+</sup> T cells were isolated and activated in vitro with anti-CD3/CD28. Following 72 h of co-culture with hUC-MSCs, CD4<sup>+</sup> T cell viability was assessed using the CCK-8 assay. The oxygen consumption rate (OCR) and glycolytic proton efflux rate (glycoPER) were measured with a Seahorse analyzer. Cytokine levels were detected by multiplex assay, and transcriptome sequencing was performed. Western blotting analyzed glucose metabolism-related enzymes and signaling pathways in lupus model mice.</p><p><strong>Results: </strong>Compared to healthy controls, activated CD4<sup>+</sup> T cells from SLE patients exhibited significantly increased OCR and glycoPER levels (P < 0.05). Following 72 h of co-culture with hUC-MSCs, OCR, glycoPER, cell viability, and pro-inflammatory factors in SLE-CD4<sup>+</sup> T cells decreased markedly (P < 0.01). Upregulation of 434 genes and downregulation of 172 genes was observed, particularly in the JAK-STAT and PI3K-Akt pathways. hUC-MSCs inhibited the expression of glucose metabolism-related enzymes and the JAK-STAT and PI3K-Akt signaling pathways in lupus model mice.</p><p><strong>Conclusion: </strong>hUC-MSCs inhibited the proliferation and function of aberrant CD4<sup>+</sup> T cells in SLE patients by modulating glycometabolism and the JAK-STAT and PI3K-Akt signaling pathways, providing new insights into the therapeutic mechanisms of MSCs based on metabolic reprogramming.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143459643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stem Cell-Derived Extracellular Vesicles for Acute Pancreatitis: a Systematic Review and Meta-analysis of Preclinical Studies.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-02-14 DOI: 10.1007/s12015-025-10852-5
Yinghui Hong, Mingliang Ye, Junshi Wang, Lei Huang

Background: Several studies have reported the effectiveness of stem cell-derived extracellular vesicles (SC-EVs) in disease treatment. However, the efficacy of SC-EVs for severe acute pancreatitis (SAP) remains uncertain. This systematic review aimed to analyze and evaluate the effect of SC-EVs in the treatment of SAP in animal models by summarizing data from published studies.

Methods: We searched Pubmed, Embase, and Web of Science databases to identify preclinical studies investigating the therapeutic effect of SC-EVs on SAP. The primary outcome was the histopathological scores of pancreatic tissues, including inflammation, edema, and necrosis. Other outcome measures included levels of amylase, IL-6, IL-10, and TNF-α. Eligible studies were selected based on the inclusion and exclusion criteria. SYRCLE checklist was adopted to assess the quality and bias risks of included studies. Mean differences and 95% confidence intervals were calculated using the inverse variance method with a random effects model. All statistical analyses were performed using RevMan 5.3 software.

Results: A total of 8 studies including 126 animals were included. The results of meta-analysis revealed that SC-EVs treatment significantly reduced pancreatic histopathologic scores (total score: MD = -5.17, 95% CI: -5.79, -4.55; inflammation score: MD = -1.44, 95% CI: -1.70, -1.19; edema score: MD = -1.42, 95% CI: -1.75, -1.09; necrosis score: MD = -1.42, 95% CI: -1.80, -1.04), inhibited pro-inflammatory factor release (IL-6: SMD = -3.20, 95% CI: -4.51, -1.88; TNF-α SMD = -5.18, 95% CI: -6.96, -3.40), and enhancing the release of anti-inflammatory factors (IL-10 SMD = 4.15, 95% CI: 2.49, 5.81). Further subgroup analyses displayed SC-EVs treatment obviously attenuated animal pancreatic pathologic injury in traumatic pancreatitis and drug-induced acute pancreatitis, and the effect of SC-EVs to inhibit TNF-α secretion in the drug-induced SAP model was correlated with the dose of SC-EVs injection.

Conclusions: This meta-analysis displayed that SC-EVs were correlated with SAP injury alleviation and pancreas function reservation. Research into the treatment of SAP with SC-EVs is still in its early stage, necessitating further comprehensive investigations in the future to elucidate the therapeutic mechanisms of SC-EVs and their potential application in SAP.

{"title":"Stem Cell-Derived Extracellular Vesicles for Acute Pancreatitis: a Systematic Review and Meta-analysis of Preclinical Studies.","authors":"Yinghui Hong, Mingliang Ye, Junshi Wang, Lei Huang","doi":"10.1007/s12015-025-10852-5","DOIUrl":"https://doi.org/10.1007/s12015-025-10852-5","url":null,"abstract":"<p><strong>Background: </strong>Several studies have reported the effectiveness of stem cell-derived extracellular vesicles (SC-EVs) in disease treatment. However, the efficacy of SC-EVs for severe acute pancreatitis (SAP) remains uncertain. This systematic review aimed to analyze and evaluate the effect of SC-EVs in the treatment of SAP in animal models by summarizing data from published studies.</p><p><strong>Methods: </strong>We searched Pubmed, Embase, and Web of Science databases to identify preclinical studies investigating the therapeutic effect of SC-EVs on SAP. The primary outcome was the histopathological scores of pancreatic tissues, including inflammation, edema, and necrosis. Other outcome measures included levels of amylase, IL-6, IL-10, and TNF-α. Eligible studies were selected based on the inclusion and exclusion criteria. SYRCLE checklist was adopted to assess the quality and bias risks of included studies. Mean differences and 95% confidence intervals were calculated using the inverse variance method with a random effects model. All statistical analyses were performed using RevMan 5.3 software.</p><p><strong>Results: </strong>A total of 8 studies including 126 animals were included. The results of meta-analysis revealed that SC-EVs treatment significantly reduced pancreatic histopathologic scores (total score: MD = -5.17, 95% CI: -5.79, -4.55; inflammation score: MD = -1.44, 95% CI: -1.70, -1.19; edema score: MD = -1.42, 95% CI: -1.75, -1.09; necrosis score: MD = -1.42, 95% CI: -1.80, -1.04), inhibited pro-inflammatory factor release (IL-6: SMD = -3.20, 95% CI: -4.51, -1.88; TNF-α SMD = -5.18, 95% CI: -6.96, -3.40), and enhancing the release of anti-inflammatory factors (IL-10 SMD = 4.15, 95% CI: 2.49, 5.81). Further subgroup analyses displayed SC-EVs treatment obviously attenuated animal pancreatic pathologic injury in traumatic pancreatitis and drug-induced acute pancreatitis, and the effect of SC-EVs to inhibit TNF-α secretion in the drug-induced SAP model was correlated with the dose of SC-EVs injection.</p><p><strong>Conclusions: </strong>This meta-analysis displayed that SC-EVs were correlated with SAP injury alleviation and pancreas function reservation. Research into the treatment of SAP with SC-EVs is still in its early stage, necessitating further comprehensive investigations in the future to elucidate the therapeutic mechanisms of SC-EVs and their potential application in SAP.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143441987","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Improving the Wound Healing Process: Pivotal role of Mesenchymal stromal/stem Cells and Immune Cells.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-02-08 DOI: 10.1007/s12015-025-10849-0
Mahvash Sadeghi, Asma Moghaddam, Amir Mohammad Amiri, Kianush Charoghdoozi, Mojgan Mohammadi, Sajad Dehnavi, Mahmoud Orazizadeh

Wound healing, a physiological process, involves several different types of cells, from immune cells to non-immune cells, including mesenchymal stromal/stem cells (MSC), and their interactions. Immune cells including macrophages, neutrophils, dendritic cells (DC), innate lymphoid cells (ILC), natural killer (NK) cells, and B and T lymphocytes participate in wound healing by secreting various mediators and interacting with other cells. MSCs, as self-renewing, fast proliferating, and multipotent stromal/stem cells, are found in a wide variety of tissues and critically involved in different phases of wound healing by secreting various molecules that help to improve tissue healing and regeneration. In this review, first, we described the four main phases of wound healing, second, we reviewed the function of MSCs, MSC secretome and immune cells in improving the progress of wound repair (mainly focusing on skin wound healing), third, we explained the immune cells/MSCs interactions in the process of wound healing and regeneration, and finally, we introduce clinical applications of MSCs to improve the process of wound healing.

{"title":"Improving the Wound Healing Process: Pivotal role of Mesenchymal stromal/stem Cells and Immune Cells.","authors":"Mahvash Sadeghi, Asma Moghaddam, Amir Mohammad Amiri, Kianush Charoghdoozi, Mojgan Mohammadi, Sajad Dehnavi, Mahmoud Orazizadeh","doi":"10.1007/s12015-025-10849-0","DOIUrl":"https://doi.org/10.1007/s12015-025-10849-0","url":null,"abstract":"<p><p>Wound healing, a physiological process, involves several different types of cells, from immune cells to non-immune cells, including mesenchymal stromal/stem cells (MSC), and their interactions. Immune cells including macrophages, neutrophils, dendritic cells (DC), innate lymphoid cells (ILC), natural killer (NK) cells, and B and T lymphocytes participate in wound healing by secreting various mediators and interacting with other cells. MSCs, as self-renewing, fast proliferating, and multipotent stromal/stem cells, are found in a wide variety of tissues and critically involved in different phases of wound healing by secreting various molecules that help to improve tissue healing and regeneration. In this review, first, we described the four main phases of wound healing, second, we reviewed the function of MSCs, MSC secretome and immune cells in improving the progress of wound repair (mainly focusing on skin wound healing), third, we explained the immune cells/MSCs interactions in the process of wound healing and regeneration, and finally, we introduce clinical applications of MSCs to improve the process of wound healing.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143374868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimization of Transcription Factor-Driven Neuronal Differentiation from Human Induced Pluripotent Stem Cells for Disease Modelling and Drug Screening.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-31 DOI: 10.1007/s12015-025-10845-4
Martina Servetti, Martino Caramia, Giulia Parodi, Fabrizio Loiacono, Ennio Nano, Giorgia Biddau, Lorenzo Ferrando, Lisastella Morinelli, Pierluigi Valente, Sergio Martinoia, Andrea Escelsior, Gianluca Serafini, Serena Tamburro, Simona Baldassari, Anna Fassio, Fabio Benfenati, Anna Corradi, Bruno Sterlini

Progress of human brain in vitro models stands as a keystone in neurological and psychiatric research, addressing the limitations posed by species-specific differences in animal models. The generation of human neurons from induced pluripotent stem cells (iPSCs) using transcription factor reprogramming protocols has been shown to reduce heterogeneity and improve consistency across different stem cell lines. Despite notable advancements, the current protocols still exhibit several shortcomings. This study focuses on standardizing and optimizing the procedure for iPSC-derived glutamatergic neurons generation through the inducible overexpression of Neurogenin-2. Noteworthy refinements include stringent scrutiny of genomic rearrangements post-fibroblast reprogramming, selection of a homogeneously integrated NGN2-cassettes population, and the incorporation of an intermediate step during neuronal differentiation to store neuronal progenitors. The neural culture showed a high degree of neuronal maturation and consistency, as shown by single-cell and network electrophysiological recordings. These advancements aim to provide more reliable tools for disease modelling and drug screening in neurological disorders.

{"title":"Optimization of Transcription Factor-Driven Neuronal Differentiation from Human Induced Pluripotent Stem Cells for Disease Modelling and Drug Screening.","authors":"Martina Servetti, Martino Caramia, Giulia Parodi, Fabrizio Loiacono, Ennio Nano, Giorgia Biddau, Lorenzo Ferrando, Lisastella Morinelli, Pierluigi Valente, Sergio Martinoia, Andrea Escelsior, Gianluca Serafini, Serena Tamburro, Simona Baldassari, Anna Fassio, Fabio Benfenati, Anna Corradi, Bruno Sterlini","doi":"10.1007/s12015-025-10845-4","DOIUrl":"https://doi.org/10.1007/s12015-025-10845-4","url":null,"abstract":"<p><p>Progress of human brain in vitro models stands as a keystone in neurological and psychiatric research, addressing the limitations posed by species-specific differences in animal models. The generation of human neurons from induced pluripotent stem cells (iPSCs) using transcription factor reprogramming protocols has been shown to reduce heterogeneity and improve consistency across different stem cell lines. Despite notable advancements, the current protocols still exhibit several shortcomings. This study focuses on standardizing and optimizing the procedure for iPSC-derived glutamatergic neurons generation through the inducible overexpression of Neurogenin-2. Noteworthy refinements include stringent scrutiny of genomic rearrangements post-fibroblast reprogramming, selection of a homogeneously integrated NGN2-cassettes population, and the incorporation of an intermediate step during neuronal differentiation to store neuronal progenitors. The neural culture showed a high degree of neuronal maturation and consistency, as shown by single-cell and network electrophysiological recordings. These advancements aim to provide more reliable tools for disease modelling and drug screening in neurological disorders.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143067202","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mitochondria Express Functional Signaling Ligand-Binding Receptors that Regulate their Biological Responses - the Novel Role of Mitochondria as Stress-Response Sentinels.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-31 DOI: 10.1007/s12015-025-10847-2
Katarzyna Brzezniakiewicz-Janus, Justyna Jarczak, Adrian Konopko, Janina Ratajczak, Magdalena Kucia, Mariusz Z Ratajczak

Evidence accumulated mitochondria, as the "powerplants of the cell," express several functional receptors for external ligands that modify their function and regulate cell biology. This review sheds new light on the role of these organelles in sensing external stimuli to facilitate energy production for cellular needs. This is possible because mitochondria express some receptors on their membranes that are responsible for their autonomous responses. This is not surprising given the widely accepted hypothesis that these intracellular organelles originated from prokaryotic ancestors that fused with eukaryotic cells during early evolution. It has been reported that mitochondria express functional estrogen, androgen, glucocorticoid, 5-hydroxytryptamine, melatonin, and cannabinoid receptors. What is intriguing is recent evidence showing that mitochondria could also be directly regulated by active mediators of intracellular complement (complosome) and intrinsic mediators of purinergic signaling. Accordingly, they express receptors for intracellular complement cleavage fragments (C5a and C3a) as well as for adenosine triphosphate (ATP), which, besides its crucial role in transferring energy in the cells, is also an important signaling molecule interacting with P2X7 receptor expressed not only on the cell surface but also on the mitochondria membrane. Based on this, intrinsic complosome and purinergic signaling mediators emerge as important cooperating regulators of reactive oxygen species (ROS) release from mitochondria and activators of intracellular pattern recognition receptor Nlrp3 inflammasome. This activation within the beneficial "hormetic zone response" regulates cell metabolism, proliferation, migration, and adaptation to the surrounding challenges of the microenvironment in a favorable way.

{"title":"Mitochondria Express Functional Signaling Ligand-Binding Receptors that Regulate their Biological Responses - the Novel Role of Mitochondria as Stress-Response Sentinels.","authors":"Katarzyna Brzezniakiewicz-Janus, Justyna Jarczak, Adrian Konopko, Janina Ratajczak, Magdalena Kucia, Mariusz Z Ratajczak","doi":"10.1007/s12015-025-10847-2","DOIUrl":"https://doi.org/10.1007/s12015-025-10847-2","url":null,"abstract":"<p><p>Evidence accumulated mitochondria, as the \"powerplants of the cell,\" express several functional receptors for external ligands that modify their function and regulate cell biology. This review sheds new light on the role of these organelles in sensing external stimuli to facilitate energy production for cellular needs. This is possible because mitochondria express some receptors on their membranes that are responsible for their autonomous responses. This is not surprising given the widely accepted hypothesis that these intracellular organelles originated from prokaryotic ancestors that fused with eukaryotic cells during early evolution. It has been reported that mitochondria express functional estrogen, androgen, glucocorticoid, 5-hydroxytryptamine, melatonin, and cannabinoid receptors. What is intriguing is recent evidence showing that mitochondria could also be directly regulated by active mediators of intracellular complement (complosome) and intrinsic mediators of purinergic signaling. Accordingly, they express receptors for intracellular complement cleavage fragments (C5a and C3a) as well as for adenosine triphosphate (ATP), which, besides its crucial role in transferring energy in the cells, is also an important signaling molecule interacting with P2X7 receptor expressed not only on the cell surface but also on the mitochondria membrane. Based on this, intrinsic complosome and purinergic signaling mediators emerge as important cooperating regulators of reactive oxygen species (ROS) release from mitochondria and activators of intracellular pattern recognition receptor Nlrp3 inflammasome. This activation within the beneficial \"hormetic zone response\" regulates cell metabolism, proliferation, migration, and adaptation to the surrounding challenges of the microenvironment in a favorable way.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143066652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stem Cell Therapy Modulates Molecular Cues of Vasogenic Edema Following Ischemic Stroke: Role of Sirtuin-1 in Regulating Aquaporin-4 Expression.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-31 DOI: 10.1007/s12015-025-10846-3
Aishika Datta, Bijoyani Ghosh, Anirban Barik, Gautam Karmarkar, Deepaneeta Sarmah, Anupom Borah, Shailendra Saraf, Dileep R Yavagal, Pallab Bhattacharya

Background: Conventional post-stroke edema management strategies are limitedly successful as in multiple cases of hemorrhagic transformation is being reported. Clinically, acute-ischemic-stroke (AIS) intervention by endovascular mesenchymal stem cells (MSCs) have shown benefits by altering various signaling pathways. Our previous studies have reported that intra-arterial administration of 1*105 MSCs (IA-MSCs) were beneficial in alleviating post-stroke edema by modulating PKCδ/MMP9/AQP4 axis and helpful in preserving the integrity of blood-brain-barrier (BBB). However, the role of mitochondrial dysfunction and ROS generation post-AIS cannot be overlooked in context to the alteration of the BBB integrity and edema formation through the activation of inflammatory pathways. The anti-inflammatory activity of IA-MSCs in stroke has been reported to be regulated by sirtuin-1 (SIRT-1). Hence, the relationship between SIRT-1 and AQP4 towards regulation of post-stroke edema needs to be further explored. Therefore, the present study deciphers the molecular events towards AQP4 upregulation, mitochondrial dysfunction and BBB disruption in context to the modulation of SIRT-1/PKCδ/NFκB loop by IA-MSCs administration.

Methods: Ovariectomized SD rats were subjected to focal ischemia. SIRT-1 activator, SIRT-1 inhibitor, NFkB inhibitor and IA-MSCs were administered at optimized dose. At 24 h of reperfusion, behavioral tests were performed, and brains were harvested following euthanasia for molecular studies.

Results: IA-MSCs downregulated AQP4, PKCδ and NFkB expression, and upregulated SIRT-1 expression. SIRT-1 upregulation renders mitochondrial protection via reduction of oxidative stress resulting in BBB protection.

Conclusion: IA-MSCs can modulate SIRT-1 mediated AQP4 expression via mitochondrial ROS reduction and modification of NFkB transcriptional regulation.

{"title":"Stem Cell Therapy Modulates Molecular Cues of Vasogenic Edema Following Ischemic Stroke: Role of Sirtuin-1 in Regulating Aquaporin-4 Expression.","authors":"Aishika Datta, Bijoyani Ghosh, Anirban Barik, Gautam Karmarkar, Deepaneeta Sarmah, Anupom Borah, Shailendra Saraf, Dileep R Yavagal, Pallab Bhattacharya","doi":"10.1007/s12015-025-10846-3","DOIUrl":"https://doi.org/10.1007/s12015-025-10846-3","url":null,"abstract":"<p><strong>Background: </strong>Conventional post-stroke edema management strategies are limitedly successful as in multiple cases of hemorrhagic transformation is being reported. Clinically, acute-ischemic-stroke (AIS) intervention by endovascular mesenchymal stem cells (MSCs) have shown benefits by altering various signaling pathways. Our previous studies have reported that intra-arterial administration of 1*10<sup>5</sup> MSCs (IA-MSCs) were beneficial in alleviating post-stroke edema by modulating PKCδ/MMP9/AQP4 axis and helpful in preserving the integrity of blood-brain-barrier (BBB). However, the role of mitochondrial dysfunction and ROS generation post-AIS cannot be overlooked in context to the alteration of the BBB integrity and edema formation through the activation of inflammatory pathways. The anti-inflammatory activity of IA-MSCs in stroke has been reported to be regulated by sirtuin-1 (SIRT-1). Hence, the relationship between SIRT-1 and AQP4 towards regulation of post-stroke edema needs to be further explored. Therefore, the present study deciphers the molecular events towards AQP4 upregulation, mitochondrial dysfunction and BBB disruption in context to the modulation of SIRT-1/PKCδ/NFκB loop by IA-MSCs administration.</p><p><strong>Methods: </strong>Ovariectomized SD rats were subjected to focal ischemia. SIRT-1 activator, SIRT-1 inhibitor, NFkB inhibitor and IA-MSCs were administered at optimized dose. At 24 h of reperfusion, behavioral tests were performed, and brains were harvested following euthanasia for molecular studies.</p><p><strong>Results: </strong>IA-MSCs downregulated AQP4, PKCδ and NFkB expression, and upregulated SIRT-1 expression. SIRT-1 upregulation renders mitochondrial protection via reduction of oxidative stress resulting in BBB protection.</p><p><strong>Conclusion: </strong>IA-MSCs can modulate SIRT-1 mediated AQP4 expression via mitochondrial ROS reduction and modification of NFkB transcriptional regulation.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143067446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
hUC-MSCs Prevent Acute High-Altitude Injury through Apoe/Pdgf-b/p-Erk1/2 Axis in Mice.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-28 DOI: 10.1007/s12015-024-10840-1
Siyu Yan, Youkun Bi, Qun Liu, Shaole Song, Lihong Ma, Guangju Ji

Background: The hypobaric hypoxic atmosphere can cause adverse reactions or sickness. The purpose of this study was to explore the preventive effect and mechanism of human umbilical cord mesenchymal stem cells (hUC-MSCs) on acute pathological injury in mice exposed to high-altitude.

Methods: We pretreated C57BL/6 mice with hUC-MSCs via the tail vein injection, and then the mice were subjected to hypobaric hypoxic conditions for five days. The effects of hUC-MSCs on the pathological injury of lung, heart, brain were assessed by biochemical analysis, histopathological testing, quantitative real-time polymerase chain reaction (qPCR), and western blot (WB). Further, transcriptome sequencing was used to screen for the potential therapeutic targets of hUC-MSCs in acute pathological injury, the identified signaling axis was characterized using Apoe-/- mice, qPCR and WB.

Results: hUC-MSCs administration notably prevented and relieved gastrointestinal symptoms and inflammation of lung and heart, increased blood oxygen saturation and serum superoxide dismutase (SOD) level, decreased serum malondialdehyde (MDA) level, rescued lung tissue injury and myocardial mitochondrial disorder, elevated nissl bodies number in brain tissue and reduced the degree of pulmonary and cerebral edema. Furthermore, hUC-MSCs pretreatment reversed the down-regulated Apoe and up-regulated Pdgf-b and p-Erk1/2 in the lung of hypobaric hypoxic mice. Thus, hUC-MSCs protected against acute pathological injury caused by hypobaric hypoxic condition via the Apoe/Pdgf-b/p-Erk1/2 axis, and the identified pathway was confirmed by the negative results of Apoe-/- mice.

Conclusion: hUC-MSCs possess the preventive effect on acute pathological injury caused by hypobaric hypoxia environment at high-altitude.

{"title":"hUC-MSCs Prevent Acute High-Altitude Injury through Apoe/Pdgf-b/p-Erk1/2 Axis in Mice.","authors":"Siyu Yan, Youkun Bi, Qun Liu, Shaole Song, Lihong Ma, Guangju Ji","doi":"10.1007/s12015-024-10840-1","DOIUrl":"https://doi.org/10.1007/s12015-024-10840-1","url":null,"abstract":"<p><strong>Background: </strong>The hypobaric hypoxic atmosphere can cause adverse reactions or sickness. The purpose of this study was to explore the preventive effect and mechanism of human umbilical cord mesenchymal stem cells (hUC-MSCs) on acute pathological injury in mice exposed to high-altitude.</p><p><strong>Methods: </strong>We pretreated C57BL/6 mice with hUC-MSCs via the tail vein injection, and then the mice were subjected to hypobaric hypoxic conditions for five days. The effects of hUC-MSCs on the pathological injury of lung, heart, brain were assessed by biochemical analysis, histopathological testing, quantitative real-time polymerase chain reaction (qPCR), and western blot (WB). Further, transcriptome sequencing was used to screen for the potential therapeutic targets of hUC-MSCs in acute pathological injury, the identified signaling axis was characterized using Apoe<sup>-/-</sup> mice, qPCR and WB.</p><p><strong>Results: </strong>hUC-MSCs administration notably prevented and relieved gastrointestinal symptoms and inflammation of lung and heart, increased blood oxygen saturation and serum superoxide dismutase (SOD) level, decreased serum malondialdehyde (MDA) level, rescued lung tissue injury and myocardial mitochondrial disorder, elevated nissl bodies number in brain tissue and reduced the degree of pulmonary and cerebral edema. Furthermore, hUC-MSCs pretreatment reversed the down-regulated Apoe and up-regulated Pdgf-b and p-Erk1/2 in the lung of hypobaric hypoxic mice. Thus, hUC-MSCs protected against acute pathological injury caused by hypobaric hypoxic condition via the Apoe/Pdgf-b/p-Erk1/2 axis, and the identified pathway was confirmed by the negative results of Apoe<sup>-/-</sup> mice.</p><p><strong>Conclusion: </strong>hUC-MSCs possess the preventive effect on acute pathological injury caused by hypobaric hypoxia environment at high-altitude.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143053545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of Cell Fate Determining Transcription Factors for Generating Brain Endothelial Cells.
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-24 DOI: 10.1007/s12015-025-10842-7
Roya Ramezankhani, Jonathan De Smedt, Burak Toprakhisar, Bernard K van der Veer, Tine Tricot, Gert Vanmarcke, Bradley Balaton, Leo van Grunsven, Massoud Vosough, Yoke Chin Chai, Catherine Verfaillie

Reliable models of the blood-brain barrier (BBB), wherein brain microvascular endothelial cells (BMECs) play a key role in maintenance of barrier function, are essential tools for developing therapeutics and disease modeling. Recent studies explored generating BMEC-like cells from human pluripotent stem cells (hPSCs) by mimicking brain-microenvironment signals or genetic reprogramming. However, due to the lack of comprehensive transcriptional studies, the exact cellular identity of most of these cells remains poorly defined. In this study we aimed to identify the most likely master transcription factors (TFs) for inducing brain endothelial cell (EC) fate and assess the transcriptomic changes following their introduction into immature ECs. Therefore, we first generated PSC-derived immature ECs by transient overexpression of the TF, ETV2. Subsequently, by performing an extensive meta-analysis of transcriptome studies of brain and non-brain ECs, 12 candidate TFs were identified, which might fate immature ECs towards cells with brain EC features. Following combinatorial overexpression of these 12 TFs tagged with unique barcodes, single cell transcriptomics identified a subset of transduced cells that resembled mid-gestational human brain ECs. Assessment of the TF barcodes present in these cells revealed significant enrichment of the TFs ZIC3, TFAP2C, TFAP2A, and DLX2. These TFs might be useful to fate PSC-EC to BMEC-like cells, which could be incorporated in human in vitro BBB models.

脑微血管内皮细胞(BMECs)在维持血脑屏障(BBB)功能方面发挥着关键作用,而可靠的血脑屏障(BBB)模型是开发治疗药物和建立疾病模型的重要工具。最近的研究探索了通过模拟脑微环境信号或基因重编程,从人多能干细胞(hPSC)中生成类脑微血管内皮细胞(BMEC)。然而,由于缺乏全面的转录研究,大多数此类细胞的确切细胞身份仍未明确。在这项研究中,我们旨在确定最有可能诱导脑内皮细胞(EC)命运的主转录因子(TFs),并评估将它们引入未成熟EC后的转录组变化。因此,我们首先通过瞬时过表达 TF ETV2 生成了源自 PSC 的未成熟 EC。随后,我们对脑和非脑EC的转录组研究进行了广泛的荟萃分析,确定了12个候选TF,它们可能会使未成熟EC趋向于具有脑EC特征的细胞。在以独特条形码标记的这12个TFs进行组合过表达后,单细胞转录组学鉴定出了一个转导细胞亚群,该亚群类似于妊娠中期的人类脑ECs。对这些细胞中存在的 TF 条形码进行评估后发现,ZIC3、TFAP2C、TFAP2A 和 DLX2 等 TF 显著富集。这些TFs可能有助于将PSC-EC转化为BMEC样细胞,并将其纳入人类体外BBB模型。
{"title":"Identification of Cell Fate Determining Transcription Factors for Generating Brain Endothelial Cells.","authors":"Roya Ramezankhani, Jonathan De Smedt, Burak Toprakhisar, Bernard K van der Veer, Tine Tricot, Gert Vanmarcke, Bradley Balaton, Leo van Grunsven, Massoud Vosough, Yoke Chin Chai, Catherine Verfaillie","doi":"10.1007/s12015-025-10842-7","DOIUrl":"https://doi.org/10.1007/s12015-025-10842-7","url":null,"abstract":"<p><p>Reliable models of the blood-brain barrier (BBB), wherein brain microvascular endothelial cells (BMECs) play a key role in maintenance of barrier function, are essential tools for developing therapeutics and disease modeling. Recent studies explored generating BMEC-like cells from human pluripotent stem cells (hPSCs) by mimicking brain-microenvironment signals or genetic reprogramming. However, due to the lack of comprehensive transcriptional studies, the exact cellular identity of most of these cells remains poorly defined. In this study we aimed to identify the most likely master transcription factors (TFs) for inducing brain endothelial cell (EC) fate and assess the transcriptomic changes following their introduction into immature ECs. Therefore, we first generated PSC-derived immature ECs by transient overexpression of the TF, ETV2. Subsequently, by performing an extensive meta-analysis of transcriptome studies of brain and non-brain ECs, 12 candidate TFs were identified, which might fate immature ECs towards cells with brain EC features. Following combinatorial overexpression of these 12 TFs tagged with unique barcodes, single cell transcriptomics identified a subset of transduced cells that resembled mid-gestational human brain ECs. Assessment of the TF barcodes present in these cells revealed significant enrichment of the TFs ZIC3, TFAP2C, TFAP2A, and DLX2. These TFs might be useful to fate PSC-EC to BMEC-like cells, which could be incorporated in human in vitro BBB models.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143034205","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Empagliflozin Reduces High Glucose-Induced Cardiomyopathy in hiPSC-Derived Cardiomyocytes : Glucose-induced Lipotoxicity in hiPSC-Derived Cardiomyocytes. 恩格列净降低高糖诱导的hipsc源性心肌细胞心肌病:葡萄糖诱导的hipsc源性心肌细胞脂毒性
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-22 DOI: 10.1007/s12015-024-10839-8
Hsiu-Hui Tsai, Fu-Chih Hsiao, Alice L Yu, Jyuhn-Huarng Juang, John Yu, Pao-Hsien Chu

Human-induced pluripotent stem cell (hiPSC) technology has been applied in pathogenesis studies, drug screening, tissue engineering, and stem cell therapy, and patient-specific hiPSC-derived cardiomyocytes (hiPSC-CMs) have shown promise in disease modeling, including diabetic cardiomyopathy. High glucose (HG) treatment induces lipotoxicity in hiPSC-CMs, as evidenced by changes in cell size, beating rate, calcium handling, and lipid accumulation. Empagliflozin, an SGLT2 inhibitor, effectively mitigates the hypertrophic changes, abnormal calcium handling, and contractility impairment induced by HG. Glucose concentration influences SGLT2 expression in cardiomyocytes, highlighting its potential role in diabetic cardiomyopathy. These findings support the potential utility of hiPSC-CMs in studying diabetic cardiomyopathy and the efficacy of empagliflozin in ameliorating HG-induced cardiomyocyte dysfunction. Such research may advance developments in precision medicine and therapeutic interventions for patients with diabetic cardiomyopathy.

人诱导多能干细胞(hiPSC)技术已被应用于发病机制研究、药物筛选、组织工程和干细胞治疗,患者特异性hiPSC来源的心肌细胞(hiPSC- cms)在疾病建模,包括糖尿病心肌病中显示出前景。高糖(HG)处理诱导hiPSC-CMs中的脂肪毒性,这可以通过细胞大小、跳动速率、钙处理和脂质积累的变化来证明。恩格列清是一种SGLT2抑制剂,可有效减轻HG引起的肥厚变化、钙处理异常和收缩性损伤。葡萄糖浓度影响心肌细胞中SGLT2的表达,突出其在糖尿病性心肌病中的潜在作用。这些发现支持hiPSC-CMs在研究糖尿病心肌病和恩格列净改善hg诱导的心肌细胞功能障碍方面的潜在效用。这些研究可能会促进糖尿病心肌病患者的精准医学和治疗干预的发展。
{"title":"Empagliflozin Reduces High Glucose-Induced Cardiomyopathy in hiPSC-Derived Cardiomyocytes : Glucose-induced Lipotoxicity in hiPSC-Derived Cardiomyocytes.","authors":"Hsiu-Hui Tsai, Fu-Chih Hsiao, Alice L Yu, Jyuhn-Huarng Juang, John Yu, Pao-Hsien Chu","doi":"10.1007/s12015-024-10839-8","DOIUrl":"https://doi.org/10.1007/s12015-024-10839-8","url":null,"abstract":"<p><p>Human-induced pluripotent stem cell (hiPSC) technology has been applied in pathogenesis studies, drug screening, tissue engineering, and stem cell therapy, and patient-specific hiPSC-derived cardiomyocytes (hiPSC-CMs) have shown promise in disease modeling, including diabetic cardiomyopathy. High glucose (HG) treatment induces lipotoxicity in hiPSC-CMs, as evidenced by changes in cell size, beating rate, calcium handling, and lipid accumulation. Empagliflozin, an SGLT2 inhibitor, effectively mitigates the hypertrophic changes, abnormal calcium handling, and contractility impairment induced by HG. Glucose concentration influences SGLT2 expression in cardiomyocytes, highlighting its potential role in diabetic cardiomyopathy. These findings support the potential utility of hiPSC-CMs in studying diabetic cardiomyopathy and the efficacy of empagliflozin in ameliorating HG-induced cardiomyocyte dysfunction. Such research may advance developments in precision medicine and therapeutic interventions for patients with diabetic cardiomyopathy.</p>","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phenotypic Characterisation of Bone Marrow-Derived Haematopoietic Stem/Progenitor Cells from HIV-Infected Individuals. hiv感染者骨髓来源造血干细胞/祖细胞的表型特征
IF 4.5 3区 医学 Q2 CELL & TISSUE ENGINEERING Pub Date : 2025-01-21 DOI: 10.1007/s12015-024-10834-z
Priyal Mistry, Joachim J C Potgieter, Michael S Pepper, Chrisna Durandt
{"title":"Phenotypic Characterisation of Bone Marrow-Derived Haematopoietic Stem/Progenitor Cells from HIV-Infected Individuals.","authors":"Priyal Mistry, Joachim J C Potgieter, Michael S Pepper, Chrisna Durandt","doi":"10.1007/s12015-024-10834-z","DOIUrl":"https://doi.org/10.1007/s12015-024-10834-z","url":null,"abstract":"","PeriodicalId":21955,"journal":{"name":"Stem Cell Reviews and Reports","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011928","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Stem Cell Reviews and Reports
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1