Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Front Immunol ; 14: 1136290, 2023.
Article in English | MEDLINE | ID: mdl-37275899

ABSTRACT

The mortality rate associated with acute lung injury (ALI) and its severe form, acute respiratory distress syndrome, is high. Induced pluripotent stem cell (iPSC) therapy is a potential treatment method for ALI, but its therapeutic efficacy is limited in injured lungs. Nitric oxide (NO) has various physiological actions. The current study investigated the effect of iPSCs pretreated with NO donors in paraquat (PQ)-induced ALI mouse model. Male C57BL/6 mice were intraperitoneally injected with PQ, followed by infusion of phosphate-buffered saline, iPSCs, L-arginine pretreated iPSCs, or Nitro-L-arginine methylester (L-NAME) pretreated iPSCs through the tail veins. Histopathological changes, pulmonary microvascular permeability, and inflammatory cytokine levels were analyzed after 3 or 28 d. The effects on iPSC proliferation, migration, and adhesion were evaluated in vitro. More L-arginine-pretreated iPSCs were selectively trafficked into the injured pulmonary tissue of mice with LPS-induced ALI, drastically diminishing the histopathologic changes and inflammatory cytokine levels (IL-1ß and IL-6). There was also markedly improved pulmonary microvascular permeability and pulmonary function. The NO inhibitor abolished the protective effects of iPSCs. In addition, the ability of L-arginine to promote the proliferation and migration of iPSCs was decreased by L-NAME pretreatment, suggesting that NO might mediate the therapeutic benefits of iPSC. The improvement of the iPSC physiological changes by the endogenous gaseous molecule NO reduces lung injury severity. L-Arginine represents a pharmacologically important strategy for enhancing the therapeutic potential of iPSCs.


Subject(s)
Acute Lung Injury , Induced Pluripotent Stem Cells , Nitric Oxide/metabolism , Paraquat/toxicity , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Mice, Inbred C57BL , Male , Animals , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Arginine/therapeutic use
2.
Aging (Albany NY) ; 15(12): 5854-5872, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37367945

ABSTRACT

Heterochronic parabiosis has shown that aging individuals can be rejuvenated by a youthful circulatory system; however, the underlying mechanisms remain unclear. Here, we evaluated the effect of exosomes isolated from mouse induced pluripotent stem cells (iPSCs) on angiogenesis in naturally aged mice. To achieve this, the angiogenic capacity of aortic ring, the total antioxidant capacity (TAOC), p53 and p16 expression levels of major organs, the proliferation of adherent bone marrow cells, and the function and content of serum exosomes in aged mice administered iPSC-derived exosomes were examined. Additionally, the effect of iPSC-derived exosomes on injured human umbilical vein endothelial cells (HUVECs) was assessed. The angiogenic capacity of aortic rings and clonality of bone marrow cells from young mice were significantly higher than those from aged mice; moreover, the organs of aged mice had a higher expression of aging genes and lower total TAOC. However, in vitro and in vivo experiments showed that the administration of iPSC-derived exosomes significantly improved these parameters in aged mice. The synergistic effect of both in vivo and in vitro treatments of aortic rings with iPSC-derived exosomes improved the angiogenic capacity of aortic rings from aged mice to levels similar to that of young mice. Compared with untreated aged mice, serum exosomal protein content and their promoted effect on endothelial cell proliferation and angiogenesis were significantly higher in untreated young mice and aged mice treated with iPSC-derived exosomes. Overall, these results showed that iPSC-derived exosomes may rejuvenate the body by anti-aging the vascular system.


Subject(s)
Exosomes , Induced Pluripotent Stem Cells , MicroRNAs , Humans , Mice , Animals , Induced Pluripotent Stem Cells/metabolism , Exosomes/metabolism , Neovascularization, Physiologic , Human Umbilical Vein Endothelial Cells/metabolism , Cardiovascular Physiological Phenomena , Cell Proliferation , MicroRNAs/metabolism
3.
Stem Cell Res Ther ; 13(1): 389, 2022 07 30.
Article in English | MEDLINE | ID: mdl-35908001

ABSTRACT

BACKGROUND: Induced pluripotent stem cells (iPSCs) have the potential to promote wound healing; however, their adhesion to the extracellular matrix (ECM) might decrease iPSC migration, thereby limiting their therapeutic potential. Integrin ß1 (Itgb1) is the major integrin subunit that mediates iPSC-ECM adhesion, suggesting that knocking out Itgb1 might be an effective method for enhancing the therapeutic efficacy of iPSCs. METHODS: We knocked out Itgb1 in mouse iPSCs and evaluated its effects on the therapeutic potential of topically applied iPSCs, as well as their underlying in vivo and in vitro mechanisms. RESULTS: The Itgb1-knockout (Itgb1-KO) did not change iPSC pluripotency, function, or survival in the absence of embedding in an ECM gel but did accelerate wound healing, angiogenesis, blood perfusion, and survival in skin-wound lesions. However, embedding in an ECM gel inhibited the in vivo effects of wild-type iPSCs but not those of Itgb1-knockout iPSCs. Additionally, in vitro results showed that Itgb1-knockout decreased iPSC-ECM adhesion while increasing ECM-crossing migration. Moreover, ECM coating on the culture surface did not change cell survival, regardless of Itgb1 status; however, the in vivo and in vitro functions of both Itgb1-knockout and wild-type iPSCs were not affected by the presence of agarose gel, which does not contain integrin-binding sites. Knockout of Integrin α4 (Itga4) did not change the above-mentioned cellular and therapeutic functions of iPSCs. CONCLUSIONS: Itgb1-knockout increased iPSCs migration and the wound-healing-promoting effect of topically applied iPSCs. These findings suggest the inhibition of Itgb1 expression is a possible strategy for increasing the efficacy of iPSC therapies.


Subject(s)
Induced Pluripotent Stem Cells , Integrin beta1/metabolism , Animals , Cell Movement , Extracellular Matrix/physiology , Integrin beta1/genetics , Integrins , Mice , Mice, Knockout , Wound Healing/genetics
4.
Inorg Chem ; 61(21): 8135-8143, 2022 May 30.
Article in English | MEDLINE | ID: mdl-35588219

ABSTRACT

A series of theoretical methods, including density functional theory, multiconfiguration molecular orbital theory, and ab initio valence bond theory, are devoted to understanding the metal-ligand bonds in M-BP (BP = biphenyl; M = Sc, Y, or La) complexes. Different from most transition metal-BP complexes, the most stable metal-biphenyl conformers are not half-sandwich but clamshell. Energy decomposition analysis results reveal that the M-BP bonds in the clamshell conformers possess extra-large orbital relaxation. According to the wave function analysis, 2-fold donations and 2-fold back-donations exist in the clamshell M-BP bonds. The back-donations from M to BP are quite strong, while donations from BP to M are quite weak. Our work improves our understanding of the metal-ligand bonds, which can be considered as the "reversed" Dewar-Chatt-Duncanson model.

6.
EBioMedicine ; 71: 103575, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34488017

ABSTRACT

BACKGROUND: Human sinoatrial cardiomyocytes are essential building blocks for cell therapies of conduction system disorders. However, current differentiation protocols for deriving nodal cardiomyocytes from human pluripotent stem cells (hPSCs) are very inefficient. METHODS: By employing the hPSCs to cardiomyocyte (CM) in vitro differentiation system and generating E2A-knockout hESCs using CRISPR/Cas9 gene editing technology, we analyze the functions of E2A in CM differentiation. FINDINGS: We found that knockout of the transcription factor E2A substantially increased the proportion of nodal-like cells in hESC-derived CMs. The E2A ablated CMs displayed smaller cell size, increased beating rates, weaker contractile force, and other functional characteristics similar to sinoatrial node (SAN) cells. Transcriptomic analyses indicated that ion channel-encoding genes were up-regulated in E2A ablated CMs. E2A directly bounded to the promoters of genes key to SAN development via conserved E-box motif, and promoted their expression. Unexpect enhanced activity of NOTCH pathway after E2A ablation could also facilate to induct ventricle workingtype CMs reprogramming into SAN-like cells. INTERPRETATION: Our study revealed a new role for E2A during directed cardiac differentiation of hESCs and may provide new clues for enhancing induction efficiency of SAN-like cardiomyocytes from hPSCs in the future. FUNDING: This work was supported by the NSFC (No.82070391, N.S.; No.81870175 and 81922006, P.L.), the National Key R&D Program of China (2018YFC2000202, N.S.; 2017YFA0103700, P.L.), the Haiju program of National Children's Medical Center EK1125180102, and Innovative research team of high-level local universities in Shanghai and a key laboratory program of the Education Commission of Shanghai Municipality (ZDSYS14005).


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Action Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Human Embryonic Stem Cells/metabolism , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, SCID , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
8.
Nucleic Acids Res ; 49(4): 1972-1986, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33503260

ABSTRACT

Maintenance of stem-cell identity requires proper regulation of enhancer activity. Both transcription factors OCT4/SOX2/NANOG and histone methyltransferase complexes MLL/SET1 were shown to regulate enhancer activity, but how they are regulated in embryonic stem cells (ESCs) remains further studies. Here, we report a transcription factor BACH1, which directly interacts with OCT4/SOX2/NANOG (OSN) and MLL/SET1 methyltransferase complexes and maintains pluripotency in mouse ESCs (mESCs). BTB domain and bZIP domain of BACH1 are required for these interactions and pluripotency maintenance. Loss of BACH1 reduced the interaction between NANOG and MLL1/SET1 complexes, and decreased their occupancy on chromatin, and further decreased H3 lysine 4 trimethylation (H3K4me3) level on gene promoters and (super-) enhancers, leading to decreased enhancer activity and transcription activity, especially on stemness-related genes. Moreover, BACH1 recruited NANOG through chromatin looping and regulated remote NANOG binding, fine-tuning enhancer-promoter activity and gene expression. Collectively, these observations suggest that BACH1 maintains pluripotency in ESCs by recruiting NANOG and MLL/SET1 complexes to chromatin and maintaining the trimethylated state of H3K4 and enhancer-promoter activity, especially on stemness-related genes.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Nanog Homeobox Protein/metabolism , Promoter Regions, Genetic , Animals , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/physiology , Cell Line , Cells, Cultured , Chromatin/metabolism , Histones/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Protein Domains , SOXB1 Transcription Factors/metabolism
9.
Stem Cell Res Ther ; 11(1): 334, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32746912

ABSTRACT

BACKGROUND: Stem cells, including induced pluripotent stem cells (iPSCs), have tremendous potential in health care, though with several significant limitations. Each of the limitations, including immunogenicity, may block most of the therapeutic potentials. Beta2 microglobulin (B2M) and MHC II transactivator (CIITA) are critical for MHC I and II, respectively. MHCs are responsible for immunogenic recognition. METHODS: B2M and CIITA were knocked out from human iPSCs, either separately or simultaneously. The effects of single or dual knockout of B2M and CIITA on iPSC properties were evaluated in a xenogeneic model of human-to-monkey transplantation. RESULTS: B2M or CIITA knockout in human induced pluripotent stem cells (iPSCs) diminishes the expression of MHC I or II alleles, respectively, without changing iPSC pluripotency. Dual knockout was better than either single knockout in preserving the ability of human iPSCs to reduce infiltration of T and B lymphocytes, survive, and promote wound healing in monkey wound lesions. The knockouts did not affect the xenogeneic iPSC-induced infiltration of macrophages and natural killer cells. They, however, decreased the iPSC-promoted proliferation of allogeneic peripheral blood mononuclear cells and T lymphocytes in vitro, although not so for B lymphocytes isolated from healthy human donors. Although the dual knockout cells survived long enough for suiting therapeutic needs, the cells eventually died, possibly due to innate immune response against them, thereby eliminating long-term risks. CONCLUSIONS: Having these iPSCs with diminished immunogenicity-recognizable to allogeneic recipient may provide unlimited reproducible, universal, standardized "ready-to-use" iPSCs and relevant derivatives for clinical applications.


Subject(s)
Haplorhini , Induced Pluripotent Stem Cells , Killer Cells, Natural , Animals , Cell Line , Humans , Leukocytes, Mononuclear , Major Histocompatibility Complex
10.
Am J Transplant ; 20(10): 2755-2767, 2020 10.
Article in English | MEDLINE | ID: mdl-32277602

ABSTRACT

This study aimed to determine the mechanism of isogeneic-induced pluripotent stem cells (iPSCs) homing to vascular transplants and their therapeutic effect on chronic allogeneic vasculopathy. We found that integrin ß1 (Intgß1) was the dominant integrin ß unit in iPSCs that mediates the adhesion of circulatory and endothelial cells (ECs). Intgß1 knockout or Intgß1-siRNAs inhibit iPSC adhesion and migration across activated endothelial monolayers. The therapeutic effects of the following were examined: iPSCs, Intgß1-knockout iPSCs, iPSCs transfected with Intgß1-siRNAs or nontargeting siRNAs, iPSC-derived ECs, iPSC-derived ECs simultaneously overexpressing Intgα4 and Intgß1, iPSCs precultured in endothelial medium for 3 days (endothelial-prone stem cells), primary aortic ECs, mouse embryonic fibroblasts, and phosphate-buffered saline (control). The cells were administered every 3 days for a period of 8 weeks. iPSCs, iPSCs transfected with nontargeting siRNAs, and endothelial-prone stem cells selectively homed on the luminal surface of the allografts, differentiated into ECs, and decreased neointimal proliferation. Through a single administration, we found that iPSCs trafficked to allograft lesions, differentiated into ECs within 1 week, and survived for 4-8 weeks. The therapeutic effect of a single administration was moderate. Thus, Intgß1 and pluripotency are essential for iPSCs to treat allogeneic vasculopathy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Animals , Cell Differentiation , Endothelial Cells , Fibroblasts , Integrin beta1 , Mice
11.
Oxid Med Cell Longev ; 2020: 1609638, 2020.
Article in English | MEDLINE | ID: mdl-32308798

ABSTRACT

AIMS: Treatment for acute kidney injury (AKI) is challenging. Induced pluripotent stem cells (iPSCs) have great therapeutic potential. This study sought to determine whether iPSCs attenuate AKI and the role of reactive oxygen species (ROS). RESULTS: We intravenously injected isogenic iPSCs into mice 2 h after renal ischemia-reperfusion injury (IRI). The cells were selectively trafficked to ischemia/reperfusion-injured kidney where they decreased kidney ROS and inflammatory cytokines and improved kidney function and morphology. Pretreating the cells with ROS inhibitors before administration decreased iPSC engraftment and abolished the protective effect of iPSCs. In contrast, pretreating iPSCs with hydrogen peroxide increased iPSC engraftment and therapeutic effect. Although the intravenously administered iPSCs trafficked to the IRI kidney, the cells did not differentiate into proximal or distal tubular epithelial cells. In vitro, the capabilities of the iPSC-released substances to promote proliferation and decrease apoptosis of renal epithelial cells were increased by ROS pretreatment of iPSCs. Moreover, pretreatment of the iPSCs with ROS inhibitor had the opposite effect. Similarly, moderate concentrations of ROS increased while ROS inhibitors decreased iPSC mobility, adhesion to the extracellular matrix, and mitochondrial metabolism. Innovation and Conclusion. iPSCs decreased renal ischemia/reperfusion injury mainly through iPSC-released substances. The therapeutic effect, mitochondrial metabolism, mobility, and kidney trafficking of iPSCs were ROS dependent.


Subject(s)
Acute Kidney Injury/therapy , Induced Pluripotent Stem Cells/transplantation , Animals , Cells, Cultured , Humans , Male , Mice , Reactive Oxygen Species/metabolism
12.
J Mol Med (Berl) ; 97(6): 829-844, 2019 06.
Article in English | MEDLINE | ID: mdl-30944935

ABSTRACT

Nature of exosome-secreting cells determines exosome content and function. ALIX, involved in exosome biogenesis, promotes cell degeneration. Here, ALIX was knocked out (iPSC-ALIX-/-) and overexpressed (iPSC-ALIX3+) in induced pluripotent stem cells (iPSCs) using CRISPR-Cas9 and lentiviral transduction, respectively, and the secreted exosomes were analyzed. Exosomes from iPSC-ALIX-/- (exosome-KO), iPSC-ALIX3+ (exosome-over), and their corresponding controls contained 176, 529, 431, and 351 proteins, respectively. Exosome-over showed increased protein levels, while exosome-KO contained fewer protein types without differing in total protein content. ALIX knockout did not affect exosome uptake by endothelial cells. Exosome-over more effectively promoted cell viability than exosome-GFP, in a dose-dependent manner. All exosomes were protective for endothelial cells injured by hydrogen peroxide or cisplatin, as demonstrated by promotion of cell viability, horizontal migration, angiogenic sprouting from aortic rings, and formation of capillary-like structures, inhibition of apoptosis, and maintenance of permeability of endothelial monolayer, although exosome-over and exosome-KO had stronger and weaker effects, respectively. SNX2 was important for ALIX-mediated exosomal function. Beneficial functions of the exosomes were independent of experimental models, targeted cell types, causes of injury, exosome-producing iPSC passages, clones of ALIX knockout, and transfection batches of ALIX overexpression. Thus, we present a novel strategy to manipulate iPSCs for production of exosomes with beneficial ALIX-regulated protein composition for varied exosome functions. KEY MESSAGES: ALIX knockout and overexpression regulate protein profile in iPSC-derived exosome. ALIX knockout decreases therapeutic function of iPSC-derived exosomes. ALIX overexpression increases therapeutic function of iPSC-derived exosomes. Manipulating iPSCs can produce exosomes with more beneficial protein content.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Exosomes/metabolism , Induced Pluripotent Stem Cells/metabolism , Protective Agents/metabolism , Animals , Aorta/drug effects , Base Sequence , Calcium-Binding Proteins/deficiency , Cell Cycle Proteins/deficiency , Cell Movement/drug effects , Cisplatin/pharmacology , Endosomal Sorting Complexes Required for Transport/deficiency , Exosomes/drug effects , Exosomes/ultrastructure , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/toxicity , Induced Pluripotent Stem Cells/drug effects , Mice , Neovascularization, Physiologic/drug effects , RNA, Small Interfering/metabolism , Sorting Nexins/metabolism
13.
EBioMedicine ; 42: 443-457, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30926422

ABSTRACT

BACKGROUND: Comparing non-inbred autologous and allogeneic induced pluripotent stem cells (iPSCs) and their secreted subcellular products among non-human primates is critical for choosing optimal iPSC products for human clinical trials. METHODS: iPSCs were induced from skin fibroblastic cells of adult male rhesus macaques belonging to four unrelated consanguineous families. Teratoma generativity, host immune response, and skin wound healing promotion were evaluated subsequently. FINDINGS: All autologous, but no allogeneic, iPSCs formed teratomas, whereas all allogeneic, but no autologous, iPSCs caused lymphocyte infiltration. Macrophages were not detectable in any wound. iPSCs expressed significantly more MAMU A and E of the major histocompatibility complex (MHC) class I but not more other MHC genetic alleles than parental fibroblastic cells. All topically disseminated autologous and allogeneic iPSCs, and their exosomes accelerated skin wound healing, as demonstrated by wound closure, epithelial coverage, collagen deposition, and angiogenesis. Allogeneic iPSCs and their exosomes were less effective and viable than their autologous counterparts. Some iPSCs differentiated into new endothelial cells and all iPSCs lost their pluripotency in 14 days. Exosomes increased cell viability of injured epidermal, endothelial, and fibroblastic cells in vitro. Although exosomes contained some mRNAs of pluripotent factors, they did not impart pluripotency to host cells. INTERPRETATION: Although all of the autologous and allogeneic iPSCs and exosomes accelerated wound healing, allogeneic iPSC exosomes were the preferred choice for "off-the shelf" iPSC products, owing to their mass-production, with no concern of teratoma formation. FUND: National Natural Science Foundation of China and National Key R&D Program of China.


Subject(s)
Exosomes/metabolism , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Wound Healing/immunology , Animals , Biomarkers , Cell Survival/genetics , Cell Survival/immunology , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Macaca mulatta , Mice
14.
Acta Biomater ; 88: 540-553, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30779999

ABSTRACT

Myocardial infarction (MI) is a primary cardiovascular disease threatening human health and quality of life worldwide. The development of engineered heart tissues (EHTs) as a transplantable artificial myocardium provides a promising therapy for MI. Since most MIs occur at the ventricle, engineering ventricular-specific myocardium is therefore more desirable for future applications. Here, by combining a new macroporous 3D iron oxide scaffold (IOS) with a fixed ratio of human pluripotent stem cell (hPSC)-derived ventricular-specific cardiomyocytes and human umbilical cord-derived mesenchymal stem cells, we constructed a new type of engineered human ventricular-specific heart tissue (EhVHT). The EhVHT promoted expression of cardiac-specific genes, ion exchange, and exhibited a better Ca2+ handling behaviors and normal electrophysiological activity in vitro. Furthermore, when patched on the infarcted area, the EhVHT effectively promoted repair of heart tissues in vivo and facilitated the restoration of damaged heart function of rats with acute MI. Our results show that it is feasible to generate functional human ventricular heart tissue based on hPSC-derived ventricular myocytes for the treatment of ventricular-specific myocardium damage. STATEMENT OF SIGNIFICANCE: We successfully generated highly purified homogenous human ventricular myocytes and developed a method to generate human ventricular-specific heart tissue (EhVHT) based on three-dimensional iron oxide scaffolds. The EhVHT promoted expression of cardiac-specific genes, ion exchange, and exhibited a better Ca2+ handling behaviors and normal electrophysiological activity in vitro. Patching the EhVHT on the infarct area significantly improved cardiac function in rat acute MI models. This EhVHT has a great potential to meet the specific requirements for ventricular damages in most MI cases and for screening drugs specifically targeting ventricular myocardium.


Subject(s)
Ferric Compounds/pharmacology , Heart Ventricles/growth & development , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Differentiation/drug effects , Electrophysiological Phenomena , Heart Function Tests , Heart Ventricles/drug effects , Heart Ventricles/transplantation , Heart Ventricles/ultrastructure , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Porosity , Rats, Sprague-Dawley
15.
Theranostics ; 9(1): 290-310, 2019.
Article in English | MEDLINE | ID: mdl-30662568

ABSTRACT

Increasing evidence suggests the consensus that direct in vivo application of induced pluripotent stem cells (iPSCs) is infeasible may not be true. Methods: Teratoma formation and fate were examined in 53 normal and disease conditions involving brain, lung, liver, kidney, islet, skin, hind limb, and arteries. Results: Using classic teratoma generation assays, which require iPSCs to be congregated and confined, all mouse, human, and individualized autologous monkey iPSCs tested formed teratoma, while iPSC-derived cells did not. Intravenously or topically-disseminated iPSCs did not form teratomas with doses up to 2.5×108 iPSCs/kg and observation times up to 18 months, regardless of host tissue type; autologous, syngeneic, or immune-deficient host animals; presence or absence of disease; disease type; iPSC induction method; commercial or self-induced iPSCs; mouse, human, or monkey iPSCs; frequency of delivery; and sex. Matrigel-confined, but not PBS-suspended, syngeneic iPSCs delivered into the peritoneal cavity or renal capsule formed teratomas. Intravenously administered iPSCs were therapeutic with a dose as low as 5×106/kg and some iPSCs differentiated into somatic cells in injured organs. Disseminated iPSCs trafficked into injured tissue and survived significantly longer in injured than uninjured organs. In disease-free animals, no intravenously administered cell differentiated into an unwanted long-lasting cell or survived as a quiescent stem cell. In coculture, the stem cell medium and dominant cell-type status were critical for iPSCs to form cell masses. Conclusion: Teratoma can be easily and completely avoided by disseminating the cells. Direct in vivo iPSC application is feasible and can be safe.


Subject(s)
Cell Transplantation/adverse effects , Cell Transplantation/methods , Induced Pluripotent Stem Cells/transplantation , Teratoma/epidemiology , Animal Structures/pathology , Animals , Cells, Cultured , Disease Models, Animal , Haplorhini , Mice , Models, Theoretical , Teratoma/pathology
16.
Toxicol Lett ; 302: 60-74, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30447258

ABSTRACT

BACKGROUND: Fine ambient particle matter (PM2.5) induces inflammatory lung injury; however, whether intratracheal administration of PM2.5 increases pulmonary polymorphonuclear leukocyte (PMN) infiltration, the mechanism of infiltration, and if these cells exacerbate PM2.5-induced lung injury are unknown. METHODS: Using 32,704 subjects, the association between blood PMNs and ambient PM2.5 levels on the previous day was retrospectively analyzed. Neutropenia was achieved by injecting mice with PMN-specific antibodies. Inhibition of PMN infiltration was achieved by pretreating PMNs with soluble vascular cell adhesion molecule-1 (sVCAM-1). The effects of PMNs on PM2.5-induced lung injury and endothelial dysfunction were observed. RESULT: Short-term PM2.5 (> 75 µg/m3 air) exposure increased the PMN/white blood cell ratio and the PMN count in human peripheral blood observed during routine examination. A significant number of PM2.5-treated PMNs was able to bind sVCAM-1. In mice, intratracheally-instilled PM2.5 deposited in the alveolar space and endothelial cells, which caused significant lung edema, morphological disorder, increased permeability of the endothelial-alveolar epithelial barrier, and PMN infiltration with increased VCAM-1 expression. Depletion of circulatory PMNs inhibited these adverse effects. Replenishment of untreated PMNs, but not those pretreated with soluble VCAM-1, restored lung injury. In vitro, PM2.5 increased VCAM-1 expression and endothelial and epithelial monolayer permeability, and promoted PMN adhesion to, chemotaxis toward, and migration across these monolayers. PMNs, but not those pretreated with soluble VCAM-1, exacerbated these effects. CONCLUSION: VCAM-1-mediated PMN infiltration was essential for a detrimental cycle of PM2.5-induced inflammation and lung injury. Results suggest that drugs that inhibit PMN function might prevent acute deterioration of chronic pulmonary and cardiovascular diseases triggered by PM2.5.


Subject(s)
Lung Injury/chemically induced , Lung/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Particulate Matter , Pulmonary Edema/chemically induced , Vascular Cell Adhesion Molecule-1/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Capillary Permeability , Cell Adhesion , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Lung/immunology , Lung/pathology , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice, Inbred C57BL , Middle Aged , Neutropenia/immunology , Neutropenia/metabolism , Neutropenia/pathology , Neutrophils/immunology , Neutrophils/pathology , Particle Size , Pulmonary Edema/immunology , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Retrospective Studies , Vascular Cell Adhesion Molecule-1/immunology , Young Adult
17.
Phys Chem Chem Phys ; 20(47): 29601-29609, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30307452

ABSTRACT

New gas phase infrared spectroscopy is reported for an aromatic alpha hydroxy carboxylic acid homo dimer of 9-hydroxy-9-fluorene carboxylic acid (9HFCA)2, and the hetero dimer of 9HFCA with glycolic acid. In terms of the 9-hydroxy stretching frequency, the 16 cm-1 blue-shift in the homo dimer and the 17 cm-1 blue-shift in the hetero dimer, relative to that in 9HFCA monomer, are attributed to collective effects with anti-cooperativity stronger than cooperativity. Furthermore, for the hetero dimer, the two alpha hydroxy groups' stretching frequencies are clearly resolved, and differ by 30 cm-1. This difference represents a modest, quantitative enhancement of the intramolecular H-bond by the fluorene moiety in 9HFCA monomer, as opposed to that in glycolic acid. Accurate vibrational frequencies of the alpha OH, 3568 cm-1 in the bare glycolic acid, and 3584 cm-1 in the glycolic acid homo dimer are determined for the first time by comparison to 9HFCA monomer, homo and hetero dimers. The quantitative studies by infrared spectroscopy reveal subtle interactions among intra- and intermolecular H-bonds in the alpha hydroxyl acid dimers, which are also uniquely extended to probe each monomer's subtle intramolecular interactions.

18.
J Phys Chem A ; 122(27): 5886-5894, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-29901393

ABSTRACT

A new energy decomposition analysis (EDA) scheme based on valence bond (VB) wave function, called VB-EDA, is presented. In VB-EDA, the total interaction energy is decomposed into frozen, charge transfer, polarization and dynamic correlation terms based on valence bond calculations. The frozen term is the energy variation of the unrelaxed VB wave function according to the change of an interaction distance. The charge transfer term is the contribution of the additional VB structures while the polarization term is due to the relaxation of VB orbitals. Dynamic correlation term is computed by post-VBSCF methods. Different from other existing VB based EDA schemes, which were used to analyze noncovalent interactions for some specific complexes, the newly developed VB-EDA is designed for the general use. Using VB-EDA, the bonding nature of cation-π interactions in a series of cation-π complexes (cations = Li+, Na+, K+, Mg2+, and Ca2+; π systems = ethylene and benzene) is explored. Furthermore, a new covalency index, which demonstrates the covalency of cation-π interactions, is presented based on the VB-EDA results. The VB-EDA analysis reveals that the cation-π interactions in the Li+, Na+, and K+ complexes belong to the typical ionic bonds while the Mg2+ and Ca2+ complexes have the relatively large covalent characteristics. However, only the C2H4-Mg2+ complex can be regarded as a covalent bonding complex while the other complexes belong to the typical ionic complexes. Thereupon, it must be careful in the cognition for the covalency of intermolecular interaction. Large nonelectrostatic interaction component does not always correspond to a covalent bond.

19.
Exp Ther Med ; 15(6): 4791-4797, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29805497

ABSTRACT

Exosomes are a family of extracellular vesicles that are secreted from almost all types of cells and are associated with cell-to-cell communication. The present study was performed to investigate the effects of human induced pluripotent stem cell-derived exosomes (hiPSC-exo) on cell viability, capillary-like structure formation and senescence in endothelial cells exposed to high glucose. Exosomes were isolated from the conditional medium of hiPSCs and confirmed by transmission electron microscopy, nanoparticle tracking analysis and western blot analysis using Alix and cluster of differentiation-63 as markers. hiPSC-exo were labeled with PKH26 for tracking, and it was determined that spherical exosomes, with a typical cup-shape, were absorbed by human umbilical vascular endothelial cells (HUVECs). Cultured HUVECs were treated with high glucose (33 mM) with or without hiPSC-exo (20 µg/ml) for 48 h, and cell viability, capillary tube formation and senescence were assessed. When exposed to high glucose, viability and tube formation in HUVECs was significantly reduced (P<0.0001), whereas the proportion of senescent cells was higher compared with that in control HUVECs (P<0.0001). Furthermore, hiPSC-exo restored cell viability and capillary-like structure formation, and reduced senescence in HUVECs exposed to high glucose (P<0.0001). However, hiPSC-exo had minimal effects on normal HUVECs. These findings suggest that stem cell-derived exosomes are able to promote cell proliferation, enhance capillary-like structure formation and reduce senescence in endothelial cells exposed to high glucose.

20.
J Mol Cell Cardiol ; 117: 49-61, 2018 04.
Article in English | MEDLINE | ID: mdl-29452156

ABSTRACT

PRKAG2 cardiac syndrome is a distinct form of human cardiomyopathy characterized by cardiac hypertrophy, ventricular pre-excitation and progressive cardiac conduction disorder. However, it remains unclear how mutations in the PRKAG2 gene give rise to such a complicated disease. To investigate the underlying molecular mechanisms, we generated disease-specific hiPSC-derived cardiomyocytes from two brothers both carrying a heterozygous missense mutation c.905G>A (R302Q) in the PRKAG2 gene and further corrected the R302Q mutation with CRISPR-Cas9 mediated genome editing. Disease-specific hiPSC-cardiomyocytes recapitulated many phenotypes of PRKAG2 cardiac syndrome including cellular enlargement, electrophysiological irregularities and glycogen storage. In addition, we found that the PRKAG2-R302Q mutation led to increased AMPK activities, resulting in extensive glycogen deposition and cardiomyocyte hypertrophy. Finally we confirmed that disrupted phenotypes of PRKAG2 cardiac syndrome caused by the specific PRKAG2-R302Q mutation can be alleviated by small molecules inhibiting AMPK activity and be rescued with CRISPR-Cas9 mediated genome correction. Our results showed that disease-specific hiPSC-CMs and genetically-corrected hiPSC-cardiomyocytes would be a very useful platform for understanding the pathogenesis of, and testing autologous cell-based therapies for, PRKAG2 cardiac syndrome.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Heart Diseases/enzymology , Heart Diseases/pathology , Induced Pluripotent Stem Cells/metabolism , Models, Biological , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Adult , Base Sequence , Calcium/metabolism , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cell Differentiation , Electrophysiological Phenomena , Glycogen/metabolism , Heart Diseases/physiopathology , Humans , Male , Mitochondria/metabolism , Mutation/genetics , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Oxidation-Reduction , Phenotype , Reproducibility of Results , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...