Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Article En | MEDLINE | ID: mdl-38666423

Although significant efforts have been made in the past few decades, the development of affordable, durable, and effective electrocatalysts for direct methanol fuel cells (DMFCs) remains a formidable challenge. Herein, we present a facile and efficient phosphorization approach for synthesizing PtP2 intermetallic nanocrystals and utilize them as electrocatalysts in the methanol oxidation reaction (MOR). Impressively, the synthesized PtP2 nanocatalysts exhibit a mass activity of 2.14 mA µg-1 and a specific activity of 6.28 mA cm-2, which are 5.1 and 9.5 times higher than those achieved by the current state-of-the-art commercial Pt/C catalyst, respectively. Moreover, the PtP2 nanocatalysts demonstrate improved stability toward acidic MOR by retaining 92.1% of its initial mass activity after undergoing 5000 potential cycles, far surpassing that of the commercial Pt/C (38%). Further DMFC tests present a 2.7 times higher power density than that of the commercial Pt/C, underscoring their potential for application in methanol fuel cells. Density functional theory calculations suggest that the accelerated MOR kinetics and improved CO tolerance on PtP2 can be attributed to the attenuated binding strength of CO intermediates and the enhanced stability due to strong Pt-P interaction. To our knowledge, this is the first report identifying the MOR performance on PtP2 intermetallic nanocrystals, highlighting their potential as highly active and stable nanocatalysts for DMFCs.

2.
Stem Cell Res Ther ; 15(1): 120, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38659015

BACKGROUND: Diabetic cardiomyopathy (DCM) is a serious health-threatening complication of diabetes mellitus characterized by myocardial fibrosis and abnormal cardiac function. Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are a potential therapeutic tool for DCM and myocardial fibrosis via mechanisms such as the regulation of microRNA (miRNA) expression and inflammation. It remains unclear, however, whether hUC-MSC therapy has beneficial effects on cardiac function following different durations of diabetes and which mechanistic aspects of DCM are modulated by hUC-MSC administration at different stages of its development. This study aimed to investigate the therapeutic effects of intravenous administration of hUC-MSCs on DCM following different durations of hyperglycemia in an experimental male model of diabetes and to determine the effects on expression of candidate miRNAs, target mRNA and inflammatory mediators. METHODS: A male mouse model of diabetes was induced by multiple low-dose streptozotocin injections. The effects on severity of DCM of intravenous injections of hUC-MSCs and saline two weeks previously were compared at 10 and 18 weeks after diabetes induction. At both time-points, biochemical assays, echocardiography, histopathology, polymerase chain reaction (PCR), immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to analyze blood glucose, body weight, cardiac structure and function, degree of myocardial fibrosis and expression of fibrosis-related mRNA, miRNA and inflammatory mediators. RESULTS: Saline-treated diabetic male mice had impaired cardiac function and increased cardiac fibrosis after 10 and 18 weeks of diabetes. At both time-points, cardiac dysfunction and fibrosis were improved in hUC-MSC-treated mice. Pro-fibrotic indicators (α-SMA, collagen I, collagen III, Smad3, Smad4) were reduced and anti-fibrotic mediators (FGF-1, miRNA-133a) were increased in hearts of diabetic animals receiving hUC-MSCs compared to saline. Increased blood levels of pro-inflammatory cytokines (IL-6, TNF, IL-1ß) and increased cardiac expression of IL-6 were also observed in saline-treated mice and were reduced by hUC-MSCs at both time-points, but to a lesser degree at 18 weeks. CONCLUSION: Intravenous injection of hUC-MSCs ameliorated key functional and structural features of DCM in male mice with diabetes of shorter and longer duration. Mechanistically, these effects were associated with restoration of intra-myocardial expression of miRNA-133a and its target mRNA COL1AI as well as suppression of systemic and localized inflammatory mediators.


Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Fibrosis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , MicroRNAs , Myocardium , Umbilical Cord , Animals , Humans , Male , Mice , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/therapy , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/genetics , Fibrosis/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Myocardium/pathology , Umbilical Cord/cytology , Umbilical Cord/metabolism
3.
Stem Cell Res Ther ; 15(1): 66, 2024 Mar 05.
Article En | MEDLINE | ID: mdl-38443965

BACKGROUND AND AIMS: Mesenchymal stromal cells (MSCs) a potentially effective disease-modulating therapy for diabetic nephropathy (DN) but their clinical translation has been hampered by incomplete understanding of the optimal timing of administration and in vivo mechanisms of action. This study aimed to elucidate the reno-protective potency and associated mechanisms of single intravenous injections of human umbilical cord-derived MSCs (hUC-MSCs) following shorter and longer durations of diabetes. METHODS: A streptozotocin (STZ)-induced model of diabetes and DN was established in C57BL/6 mice. In groups of diabetic animals, human (h)UC-MSCs or vehicle were injected intravenously at 8 or 16 weeks after STZ along with vehicle-injected non-diabetic animals. Diabetes-related kidney abnormalities was analyzed 2 weeks later by urine and serum biochemical assays, histology, transmission electron microscopy and immunohistochemistry. Serum concentrations of pro-inflammatory and pro-fibrotic cytokines were quantified by ELISA. The expression of autophagy-related proteins within the renal cortices was investigated by immunoblotting. Bio-distribution of hUC-MSCs in kidney and other organs was evaluated in diabetic mice by injection of fluorescent-labelled cells. RESULTS: Compared to non-diabetic controls, diabetic mice had increases in urine albumin creatinine ratio (uACR), mesangial matrix deposition, podocyte foot process effacement, glomerular basement membrane thickening and interstitial fibrosis as well as reduced podocyte numbers at both 10 and 18 weeks after STZ. Early (8 weeks) hUC-MSC injection was associated with reduced uACR and improvements in multiple glomerular and renal interstitial abnormalities as well as reduced serum IL-6, TNF-α, and TGF-ß1 compared to vehicle-injected animals. Later (16 weeks) hUC-MSC injection also resulted in reduction of diabetes-associated renal abnormalities and serum TGF-ß1 but not of serum IL-6 and TNF-α. At both time-points, the kidneys of vehicle-injected diabetic mice had higher ratio of p-mTOR to mTOR, increased abundance of p62, lower abundance of ULK1 and Atg12, and reduced ratio of LC3B to LC3A compared to non-diabetic animals, consistent with diabetes-associated suppression of autophagy. These changes were largely reversed in the kidneys of hUC-MSC-injected mice. In contrast, neither early nor later hUC-MSC injection had effects on blood glucose and body weight of diabetic animals. Small numbers of CM-Dil-labeled hUC-MSCs remained detectable in kidneys, lungs and liver of diabetic mice at 14 days after intravenous injection. CONCLUSIONS: Single intravenous injections of hUC-MSCs ameliorated glomerular abnormalities and interstitial fibrosis in a mouse model of STZ-induced diabetes without affecting hyperglycemia, whether administered at relatively short or longer duration of diabetes. At both time-points, the reno-protective effects of hUC-MSCs were associated with reduced circulating TGF-ß1 and restoration of intra-renal autophagy.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , Kidney/abnormalities , Mesenchymal Stem Cells , Urogenital Abnormalities , Humans , Animals , Mice , Mice, Inbred C57BL , Diabetic Nephropathies/therapy , Injections, Intravenous , Transforming Growth Factor beta1 , Diabetes Mellitus, Experimental/therapy , Interleukin-6 , Tumor Necrosis Factor-alpha , Autophagy , Fibrosis , TOR Serine-Threonine Kinases
4.
Stem Cell Res Ther ; 15(1): 67, 2024 Mar 05.
Article En | MEDLINE | ID: mdl-38444003

BACKGROUND: Spinal cord injury (SCI) is a serious clinical condition that has pathological changes such as increased neuroinflammation and nerve tissue damage, which eventually manifests as fibrosis of the injured segment and the development of a spinal cord cavity leading to loss of function. Cell-based therapy, such as mesenchymal stem cells (MSCs) and neural stem cells (NSCs) are promising treatment strategies for spinal cord injury via immunological regulation and neural replacement respectively. However, therapeutic efficacy is rare reported on combined transplantation of MSC and NSC in acute mice spinal cord injury even the potential reinforcement might be foreseen. Therefore, this study was conducted to investigate the safety and efficacy of co-transplanting of MSC and NSC sheets into an SCI mice model on the locomotor function and pathological changes of injured spinal cord. METHODS: To evaluate the therapeutic effects of combination cells, acute SCI mice model were established and combined transplantation of hiPSC-NSCs and hMSCs into the lesion site immediately after the injury. Basso mouse scale was used to perform the open-field tests of hind limb motor function at days post-operation (dpo) 1, 3, 5, and 7 after SCI and every week after surgery. Spinal cord and serum samples were collected at dpo 7, 14, and 28 to detect inflammatory and neurotrophic factors. Hematoxylin-eosin (H&E) staining, masson staining and transmission electron microscopy were used to evaluate the morphological changes, fibrosis area and ultrastructure of the spinal cord. RESULT: M&N transplantation reduced fibrosis formation and the inflammation level while promoting the secretion of nerve growth factor and brain-derived neurotrophic factor. We observed significant reduction in damaged tissue and cavity area, with dramatic improvement in the M&N group. Compared with the Con group, the M&N group exhibited significantly improved behaviors, particularly limb coordination. CONCLUSION: Combined transplantation of hiPSC-NSC and hMSC could significantly ameliorate neuroinflammation, promote neuroregeneration, and decrease spinal fibrosis degree in safe and effective pattern, which would be indicated as a novel potential cell treatment option.


Induced Pluripotent Stem Cells , Spinal Cord Injuries , Animals , Mice , Neuroinflammatory Diseases , Spinal Cord Injuries/therapy , Disease Models, Animal , Fibrosis
5.
J Alzheimers Dis ; 97(3): 1007-1031, 2024.
Article En | MEDLINE | ID: mdl-38306054

Apolipoprotein E4 (APOE4), although yet-to-be fully understood, increases the risk and lowers the age of onset of Alzheimer's disease (AD), which is the major cause of dementia among elderly individuals. The endosome-lysosome and autophagy pathways, which are necessary for homeostasis in both neurons and glia, are dysregulated even in early AD. Nonetheless, the contributory roles of these pathways to developing AD-related pathologies in APOE4 individuals and models are unclear. Therefore, this review summarizes the dysregulations in the endosome-lysosome and autophagy pathways in APOE4 individuals and non-human models, and how these anomalies contribute to developing AD-relevant pathologies. The available literature suggests that APOE4 causes endosomal enlargement, increases endosomal acidification, impairs endosomal recycling, and downregulates exosome production. APOE4 impairs autophagy initiation and inhibits basal autophagy and autophagy flux. APOE4 promotes lysosome formation and trafficking and causes ApoE to accumulate in lysosomes. APOE4-mediated changes in the endosome, autophagosome and lysosome could promote AD-related features including Aß accumulation, tau hyperphosphorylation, glial dysfunction, lipid dyshomeostasis, and synaptic defects. ApoE4 protein could mediate APOE4-mediated endosome-lysosome-autophagy changes. ApoE4 impairs vesicle recycling and endosome trafficking, impairs the synthesis of autophagy genes, resists being dissociated from its receptors and degradation, and forms a stable folding intermediate that could disrupt lysosome structure. Drugs such as molecular correctors that target ApoE4 molecular structure and enhance autophagy may ameliorate the endosome-lysosome-autophagy-mediated increase in AD risk in APOE4 individuals.


Alzheimer Disease , Humans , Aged , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Alleles , Endosomes/metabolism , Lysosomes/metabolism , Autophagy/genetics
6.
Mol Neurobiol ; 2023 Dec 21.
Article En | MEDLINE | ID: mdl-38127186

C9orf72 genetic mutation is the most common genetic cause of ALS/FTD accompanied by abnormal protein insufficiency. Induced pluripotent stem cell (iPSC)-derived two-dimensional (2D) and three-dimensional (3D) cultures are providing new approaches. Therefore, this study established neuronal cell types and generated spinal cord organoids (SCOs) derived from C9orf72 knockdown human iPSCs to model ALS disease and screen the unrevealed phenotype. Wild-type (WT) iPSC lines from three healthy donor fibroblasts were established, and pluripotency and differentiation ability were identified by RT-PCR, immunofluorescence and flow cytometry. After infection by the lentivirus with C9orf72-targeting shRNA, stable C9-knockdown iPSC colonies were selected and differentiated into astrocytes, motor neurons and SCOs. Finally, we analyzed the extracted RNA-seq data of human C9 mutant/knockout iPSC-derived motor neurons and astrocytes from the GEO database and the inflammatory regulation-related genes in function and pathways. The expression of inflammatory factors was measured by qRT-PCR. The results showed that both WT-iPSCs and edited C9-iPSCs maintained a similar ability to differentiate into the three germ layers, astrocytes and motor neurons, forming SCOs in a 3D culture system. The constructed C9-SCOs have features of spinal cord development and multiple neuronal cell types, including sensory neurons, motor neurons, and other neurons. Based on the bioinformatics analysis, proinflammatory factors were confirmed to be upregulated in C9-iPSC-derived 2D cells and 3D cultured SCOs. The above differentiated models exhibited low C9orf72 expression and the pathological characteristics of ALS, especially neuroinflammation.

7.
Stem Cell Res ; 72: 103201, 2023 10.
Article En | MEDLINE | ID: mdl-37856927

In this study, fibroblasts were harvested and isolated from a healthy 14-year-old male donor and reprogrammed with four Yamanaka factors containing Oct3/4, Sox2, Klf4 and c-Myc to generate human induced pluripotent stem cell (iPSC) lines. The resulting iPSCs were integration-free, expressed normal karyotype, displayed pluripotency markers, and have been demonstrated to differentiate into cells with three germ layer. And the iPSCs were further differentiated to chondrosphere in vivo. The models could be used to test multiple differentiation protocols and also as a control for screening drugs and studying cartilage related disease.


Induced Pluripotent Stem Cells , Male , Humans , Adolescent , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation , Fibroblasts/metabolism , Cells, Cultured , Proto-Oncogene Proteins c-myc/genetics , Cellular Reprogramming
8.
Stem Cell Res ; 69: 103116, 2023 06.
Article En | MEDLINE | ID: mdl-37178573

The ε4 allele of the lipoprotein E gene (APOE4) is the strongest genetic risk factor associated with sporadic Alzheimer's disease (sAD). While the neuronal cell type-specific function of APOE4 in connection with AD pathology remains understudied. Therefore, we generated an induced pluripotent stem cells (iPSCs) line from a 77-year-old female donor with ApoE4 genetic background. We reprogrammed peripheral blood mononuclear cells (PBMCs) with non-integrative Sendai viral vectors containing reprogramming factors. Established iPSCs showed the capability of pluripotency, three-germ differentiation in vitro with normal karyotype. Hence, the generated iPSC could be a powerful tool to conduct further studies of AD mechanisms.


Alzheimer Disease , Induced Pluripotent Stem Cells , Female , Humans , Aged , Induced Pluripotent Stem Cells/metabolism , Cellular Reprogramming , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Leukocytes, Mononuclear/metabolism , Cell Line , Karyotype , Cell Differentiation/physiology
9.
Stem Cell Res ; 68: 103052, 2023 04.
Article En | MEDLINE | ID: mdl-36870256

Sporadic Alzheimer's disease (sAD) is the most common neurodegenerative disease worldwide, which is characterized by the progressive cognitive dysfunction and behavioral impairment. Here, we generated a human induced pluripotent stem cell (iPSC) line from the peripheral blood mononuclear cells (PBMCs) isolated from a 78-year-old male patient clinically diagnosed with sAD. The iPSC line expressed pluripotency markers, showed normal karyotype, and had the ability to differentiate into three germ layers in vitro. This iPSC line may provide a powerful tool for modeling AD in vitro and studying the pathogenesis of sAD.


Alzheimer Disease , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Male , Humans , Aged , Induced Pluripotent Stem Cells/metabolism , Alzheimer Disease/pathology , Neurodegenerative Diseases/metabolism , Leukocytes, Mononuclear/metabolism , Germ Layers/metabolism , Cell Differentiation
10.
Front Chem ; 11: 1122333, 2023.
Article En | MEDLINE | ID: mdl-36793765

Metal phosphides with multi-element components and amorphous structure represent a novel kind of electrocatalysts for promising activity and durability towards the oxygen evolution reaction (OER). In this work, a two-step strategy, including alloying and phosphating processes, is reported to synthesize trimetallic amorphous PdCuNiP phosphide nanoparticles for efficient OER under alkaline conditions. The synergistic effect between Pd, Cu, Ni, and P elements, as well as the amorphous structure of the obtained PdCuNiP phosphide nanoparticles, would boost the intrinsic catalytic activity of Pd nanoparticles towards a wide range of reactions. These obtained trimetallic amorphous PdCuNiP phosphide nanoparticles exhibit long-term stability, nearly a 20-fold increase in mass activity toward OER compared with the initial Pd nanoparticles, and 223 mV lower in overpotential at 10 mA cm-2. This work not only provides a reliable synthetic strategy for multi-metallic phosphide nanoparticles, but also expands the potential applications of this promising class of multi-metallic amorphous phosphides.

11.
Oxid Med Cell Longev ; 2022: 6483582, 2022.
Article En | MEDLINE | ID: mdl-36046683

Background: Astrocytes can be involved in motor neuron toxicity in amyotrophic lateral sclerosis (ALS) induced by noncell autonomous effects, and inflammatory cytokines may play the main role in mediating this process. However, the etiology of aberrant cytokine secretion is unclear. The present study assessed possible involvement of the mTOR-autophagy pathway in aberrant cytokine secretion by ALS patient iPSC-derived astrocytes. Method and Results. PBMCs from sporadic ALS patients and control subjects were reprogrammed into iPSCs, which were then differentiated into astrocytes and/or motor neurons. Comparison with control astrocytes indicated that conditioned medium of ALS astrocytes reduced the viability of the control motor neurons (p < 0.05) assessed using the MTT assay. The results of ELISA showed that the concentrations of TNFα, IL1ß, and IL6 in cell culture medium of ALS astrocytes were increased (p < 0.05). ALS astrocytes had higher p62 and mTOR levels and lower LC3BII/LC3BI ratio and ULK1 and p-Beclin-1 (Ser15) levels (p < 0.05), indicating defective autophagy. Exogenous inhibition of the mTOR-autophagy pathway, but not the activation of the pathway in control subject astrocytes, increased the levels of p62 and mTOR and concentration of IL-1ß, TNF-α, and IL-6 in cell culture medium and decreased the LC3BII/LC3BI ratio and levels of ULK1 and p-Beclin-1 (Ser15), and these changes were comparable to those in ALS astrocytes. After 48 h of rapamycin (autophagy activator) and 3-methyladenine (autophagy inhibitor) treatments, the exogenous activation of the mTOR-autophagy pathway, but not inhibition of the pathway, in ALS astrocytes significantly reduced the concentrations of TNFα, IL1ß, and IL6 in cell culture medium and reduced the levels of p62, while increasing the levels of LC3B-II/LC3B-I, ULK1, and p-Beclin-1 (Ser15), and these changes were comparable to those in control subject astrocytes. Conclusion: Alteration in the mTOR/ULK1/Beclin-1 pathway regulated cytokine secretion in ALS astrocytes, which was able to lead to noncell autonomous toxicity. Autophagy activation mitigated cytokine secretion by ALS astrocytes.


Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Autophagy , Beclin-1/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Interleukin-6/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Stem Cell Res ; 63: 102840, 2022 08.
Article En | MEDLINE | ID: mdl-35717858

Apolipoprotein E (APOE) gene encodes three protein isoforms (APOEε-22/ε-33/ε-44), which governs the transportation and metabolism of lipoproteins differently. While abnormalities in lipid and lipoprotein metabolism have been identified as risk factors for type 2 diabetes mellitus (T2DM). APOE gene polymorphisms might be correlated with increased risk of T2DM. Therefore, we presented an iPSC line harboring an APOE-ε3/ε3 alleles, a male donor suffering from T2DM combined with Hypertension. The derived iPSCs showed pluripotency, the capacity to differentiate into three germ layers, and normal karyotypes. Collectively, the present study provides a useful resource to reveal the associated mechanism of APOE isoform and T2DM.


Diabetes Mellitus, Type 2 , Induced Pluripotent Stem Cells , Alleles , Apolipoprotein E3/genetics , Apolipoproteins E/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Genotype , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged
13.
Stem Cell Res ; 61: 102746, 2022 05.
Article En | MEDLINE | ID: mdl-35305470

The ε4 allele of the Apolipoprotein E gene (APOE4) continues to be the strongest genetic risk factor associated with sporadic Alzheimer's disease since its discovery compared to the most common ε3 allele. Nevertheless, there is a lack of APOE4-mutant human neuronal models in vitroor in vivo. Hence, we presented an iPSC line of an APOE-ε4/ε4 alleles carrier, a male donor suffering from Alzheimer's disease combined with cerebral infarction. The established iPSC line showed standard characteristics of pluripotency. Collectively, the present study provides a useful resource to reveal the phenotype and explore the mechanism of APOE4 related Alzheimer's disease.


Alzheimer Disease , Induced Pluripotent Stem Cells , Alleles , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Apolipoprotein E4/genetics , Apolipoproteins E/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Middle Aged
14.
Stem Cell Res ; 57: 102613, 2021 Dec 01.
Article En | MEDLINE | ID: mdl-34871962

In this study, skin biopsy was, and the fibroblasts were isolated from the dermal explant cultures. Human induced pluripotent stem cell (iPSC) line was generated from the skin fibroblasts collected from a healthy 50-year old male donor with informed consent. The reprogramming of fibroblasts was performed with four Yamanaka factors containing Oct3/4, Sox2, Klf4 and c-Myc. The generated iPSCs were confirmed integration-free, expressed pluripotency markers, displayed the normal karyotype, and demonstrated trilineage differentiation potential. This iPSC model can be used to model physiological processes and screen drug validation in vitro.

15.
Stem Cell Res ; 54: 102450, 2021 07.
Article En | MEDLINE | ID: mdl-34218115

Apolipoprotein E ε4 allele (APOE4) is a minor allele of the APOE gene associated with a higher risk for Alzheimer's Disease (AD) and Vascular Dementia (VD). While lipid deposition and chronic inflammation in glia are the commonalities between atherosclerosis, VD, and AD. Hence, we presented an iPSC line of an AD male donor suffering from Cerebrovascular Atherosclerosis with APOE-ε4/ε4 alleles background. Furthermore, we differentiated the iPSCs into astrocyte to explore pathogenesis in APOE4 related dementia. The characterized iPSC line expressed typical pluripotency markers and showed differentiation potential and normal karyotype.


Alzheimer Disease , Induced Pluripotent Stem Cells , Alleles , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Genotype , Humans , Male
16.
Stem Cell Res ; 53: 102395, 2021 05.
Article En | MEDLINE | ID: mdl-34088020

Apolipoprotein E (ApoE) is a lipid-binding protein with ε2, ε3, and ε4 allelic variants in human. The ε4 isoform (ApoE4) is the strongest genetic risk factor for the late-onset form of Alzheimer's disease (AD), and is also associated with multiple neurological disorders, multiple sclerosis, and cerebrovascular disease. Here, induced pluripotent stem cells were derived from the peripheral blood mononuclear cells of a 70-year-old male donor with APOE-ε4/ε4 alleles background to explore pathogenesis and screen potential treatment methods in neurodegenerative diseases. In the newly-developed induced pluripotent stem cell line, the pluripotent markers were well expressed. In addition, the generated cells displayed a normal karyotype and have differentiation potential.


Alzheimer Disease , Induced Pluripotent Stem Cells , Aged , Alleles , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Apolipoproteins E/genetics , Genotype , Humans , Leukocytes, Mononuclear , Male
17.
Stem Cell Res ; 53: 102339, 2021 05.
Article En | MEDLINE | ID: mdl-33892291

In this study, skin biopsy was collected from a healthy 48-year old male donor with informed consent, and the fibroblasts were isolated from the dermal explant cultures. Here, a human induced pluripotent stem cell (iPSC) line was derived from the fibroblasts using the reprogramming four Yamanaka factors (Oct3/4, Sox2, Klf4, c-Myc). The generated iPSCs were integration-free, displayed the normal karyotype, expressed pluripotency markers and demonstrated trilineage differentiation potential in vitro. This iPSC model will be useful for investigating physiological processes, drug validation as well as a control in pathological mechanistic studies.


Induced Pluripotent Stem Cells , Cell Differentiation , Cellular Reprogramming , Fibroblasts , Humans , Kruppel-Like Factor 4 , Male , Middle Aged , Skin
18.
Stem Cell Res ; 53: 102324, 2021 05.
Article En | MEDLINE | ID: mdl-33839548

Potocki-Lupski syndrome (PTLS; MIM 610883) is a neurodevelopmental disorder associated with a 3.7 Mb copy number variant (CNV) duplication, locating in chromosome 17p11.2. (Soler-Alfonso et al., 2011). Here, a human induced pluripotent stem cell (iPSC) line was derived from the peripheral blood mononuclear cells of a 5-year-old child suffering Potocki-Lupski syndrome. The generated iPSCs were integration-free, had the 17p11.2 3.7 Mb CNV duplication with no additional genomic alterations, a stable karyotype, expressed pluripotency stem cell markers and could differentiate towards the three germ layers in vitro. Patient's derived iPSCs are a valuable resource for in vitro modeling of 17p11.2 microduplication induced Potocki-Lupski syndrome.


Induced Pluripotent Stem Cells , Abnormalities, Multiple , Child, Preschool , Chromosome Disorders , Chromosome Duplication , Comparative Genomic Hybridization , Humans , Leukocytes, Mononuclear
19.
Stem Cell Res Ther ; 12(1): 172, 2021 03 11.
Article En | MEDLINE | ID: mdl-33706803

BACKGROUND: Spinal cord injury (SCI) is a common disease that results in motor and sensory disorders and even lifelong paralysis. The transplantation of stem cells, such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), or subsequently generated stem/progenitor cells, is predicted to be a promising treatment for SCI. In this study, we aimed to investigate effect of human iPSC-derived neural stem cells (hiPSC-NSCs) and umbilical cord-derived MSCs (huMSCs) in a mouse model of acute SCI. METHODS: Acute SCI mice model were established and were randomly treated as phosphate-buffered saline (PBS) (control group), repaired with 1 × 105 hiPSC-NSCs (NSC group), and 1 × 105 huMSCs (MSC group), respectively, in a total of 54 mice (n = 18 each). Hind limb motor function was evaluated in open-field tests using the Basso Mouse Scale (BMS) at days post-operation (dpo) 1, 3, 5, and 7 after spinal cord injury, and weekly thereafter. Spinal cord and serum samples were harvested at dpo 7, 14, and 21. Haematoxylin-eosin (H&E) staining and Masson staining were used to evaluate the morphological changes and fibrosis area. The differentiation of the transplanted cells in vivo was evaluated with immunohistochemical staining. RESULTS: The hiPSC-NSC-treated group presented a significantly smaller glial fibrillary acidic protein (GFAP) positive area than MSC-treated mice at all time points. Additionally, MSC-transplanted mice had a similar GFAP+ area to mice receiving PBS. At dpo 14, the immunostained hiPSC-NSCs were positive for SRY-related high-mobility-group (HMG)-box protein-2 (SOX2). Furthermore, the transplanted hiPSC-NSCs differentiated into GFAP-positive astrocytes and beta-III tubulin-positive neurons, whereas the transplanted huMSCs differentiated into GFAP-positive astrocytes. In addition, hiPSC-NSC transplantation reduced fibrosis formation and the inflammation level. Compared with the control or huMSC transplanted group, the group with transplantation of hiPSC-NSCs exhibited significantly improved behaviours, particularly limb coordination. CONCLUSIONS: HiPSC-NSCs promote functional recovery in mice with acute SCI by replacing missing neurons and attenuating fibrosis, glial scar formation, and inflammation.


Induced Pluripotent Stem Cells , Neural Stem Cells , Spinal Cord Injuries , Animals , Cell Differentiation , Induced Pluripotent Stem Cells/transplantation , Mice , Neural Stem Cells/transplantation , Recovery of Function , Spinal Cord , Spinal Cord Injuries/therapy
20.
PLoS One ; 16(3): e0247662, 2021.
Article En | MEDLINE | ID: mdl-33705413

BACKGROUND: Diabetes mellitus as a chronic metabolic disease is threatening human health seriously. Although numerous clinical trials have been registered for the treatment of diabetes with stem cells, no articles have been published to summarize the efficacy and safety of mesenchymal stem cells (MSCs) in randomized controlled trials (RCTs). METHODS AND FINDINGS: The aim of this study was to systematically review the evidence from RCTs and, where possible, conduct meta-analyses to provide a reliable numerical summary and the most comprehensive assessment of therapeutic efficacy and safety with MSCs in diabetes. PubMed, Web of Science, Ovid, the Cochrane Library and CNKI were searched. The retrieval time was from establishment of these databases to January 4, 2020. Seven RCTs were eligible for analysis, including 413 participants. Meta-analysis results showed that there were no significant differences in the reduction of fasting plasma glucose (FPG) compared to the baseline [mean difference (MD) = -1.05, 95% confidence interval (CI) (-2.26,0.16), P<0.01, I2 = 94%] and the control group [MD = -0.62, 95%CI (-1.46,0.23), P<0.01, I2 = 87%]. The MSCs treatment group showed a significant decrease in hemoglobin (Hb) A1c [random-effects, MD = -1.32, 95%CI (-2.06, -0.57), P<0.01, I2 = 90%] after treatment. Additionally, HbA1c reduced more significantly in MSC treatment group than in control group [random-effects, MD = -0.87, 95%CI (-1.53, -0.22), P<0.01, I2 = 82%] at the end of follow-up. However, as for fasting C-peptide levels, the estimated pooled MD showed that there was no significant increase [MD = -0.07, 95%CI (-0.30, 0.16), P<0.01, I2 = 94%] in MSCs treatment group compared with that in control group. Notably, there was no significant difference in the incidence of adverse events between MSCs treatment group and control group [relative risk (RR) = 0.98, 95%CI (0.72, 1.32), P = 0.02, I2 = 70%]. The most commonly observed adverse reaction in the MSC treatment group was hypoglycemia (29.95%). CONCLUSIONS: This meta-analysis revealed MSCs therapy may be an effective and safe intervention in subjects with diabetes. However, due to the limited studies, a number of high-quality as well as large-scale RCTs should be performed to confirm these conclusions.


Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Glycated Hemoglobin/metabolism , Glycemic Control/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Blood Glucose/metabolism , C-Peptide/blood , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Fasting , Humans , Hypoglycemia/blood , Hypoglycemia/diagnosis , Hypoglycemia/etiology , Mesenchymal Stem Cell Transplantation/adverse effects , Randomized Controlled Trials as Topic , Treatment Outcome
...