Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Front Endocrinol (Lausanne) ; 12: 635405, 2021.
Article in English | MEDLINE | ID: mdl-34025576

ABSTRACT

Background: Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the ß-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. Methods: Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. Results: CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. Conclusions: Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Dipeptidyl Peptidase 4/biosynthesis , Integrin alpha1/biosynthesis , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Pluripotent Stem Cells/metabolism , Animals , C-Peptide/biosynthesis , Cell Differentiation , Cell Separation , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Microspheres
2.
Methods Mol Biol ; 2235: 119-125, 2021.
Article in English | MEDLINE | ID: mdl-33576973

ABSTRACT

Human pluripotent stem cells (hPSCs), either embryonic or induced, offer a plentiful platform for derivation of multiple cell types. Pericytes, generated from hPSCs, are multipotent precursors with vasculogenic features that exhibit high proliferation capability in long-term cultures. Administration of hPSC-pericytes into ischemic murine hind limb is associated with therapeutic angiogenesis and attenuation of muscle wasting. Here, we describe the protocol for derivation of large numbers of pericytes from spontaneously differentiating hPSC-embryoid bodies.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Pericytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Humans , Pericytes/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
3.
Int J Mol Sci ; 21(19)2020 09 29.
Article in English | MEDLINE | ID: mdl-33003641

ABSTRACT

The limited regenerative capacity of the injured myocardium leads to remodeling and often heart failure. Novel therapeutic approaches are essential. Induced pluripotent stem cells (iPSC) differentiated into cardiomyocytes are a potential future therapeutics. We hypothesized that organ-specific reprogramed fibroblasts may serve an advantageous source for future cardiomyocytes. Moreover, exosomes secreted from those cells may have a beneficial effect on cardiac differentiation and/or function. We compared RNA from different sources of human iPSC using chip gene expression. Protein expression was evaluated as well as exosome micro-RNA levels and their impact on embryoid bodies (EBs) differentiation. Statistical analysis identified 51 genes that were altered (p ≤ 0.05), and confirmed in the protein level, cardiac fibroblasts-iPSCs (CF-iPSCs) vs. dermal fibroblasts-iPSCs (DF-iPSCs). Several miRs were altered especially miR22, a key regulator of cardiac hypertrophy and remodeling. Lower expression of miR22 in CF-iPSCs vs. DF-iPSCs was observed. EBs treated with these exosomes exhibited more beating EBs p = 0.05. vs. control. We identify CF-iPSC and its exosomes as a potential source for cardiac recovery induction. The decrease in miR22 level points out that our CF-iPSC-exosomes are naïve of congestive heart cell memory, making them a potential biological source for future therapy for the injured heart.


Subject(s)
Exosomes/genetics , Heart Failure/therapy , Induced Pluripotent Stem Cells/metabolism , Myocardium/metabolism , Cell Differentiation/genetics , Exosomes/metabolism , Fibroblasts/metabolism , Heart/physiopathology , Heart Failure/genetics , Heart Failure/pathology , Humans , MicroRNAs/genetics , Myocardium/pathology , Myocytes, Cardiac/metabolism
4.
Stem Cell Res Ther ; 9(1): 152, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29871694

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a motor neuron (MN) disease characterized by the loss of MNs in the central nervous system. As MNs die, patients progressively lose their ability to control voluntary movements, become paralyzed and eventually die from respiratory/deglutition failure. Despite the selective MN death in ALS, there is growing evidence that malfunctional astrocytes play a crucial role in disease progression. Thus, transplantation of healthy astrocytes may compensate for the diseased astrocytes. METHODS: We developed a good manufacturing practice-grade protocol for generation of astrocytes from human embryonic stem cells (hESCs). The first stage of our protocol is derivation of astrocyte progenitor cells (APCs) from hESCs. These APCs can be expanded in large quantities and stored frozen as cell banks. Further differentiation of the APCs yields an enriched population of astrocytes with more than 90% GFAP expression (hES-AS). hES-AS were injected intrathecally into hSOD1G93A transgenic mice and rats to evaluate their therapeutic potential. The safety and biodistribution of hES-AS were evaluated in a 9-month study conducted in immunodeficient NSG mice under good laboratory practice conditions. RESULTS: In vitro, hES-AS possess the activities of functional healthy astrocytes, including glutamate uptake, promotion of axon outgrowth and protection of MNs from oxidative stress. A secretome analysis shows that these hES-AS also secrete several inhibitors of metalloproteases as well as a variety of neuroprotective factors (e.g. TIMP-1, TIMP-2, OPN, MIF and Midkine). Intrathecal injections of the hES-AS into transgenic hSOD1G93A mice and rats significantly delayed disease onset and improved motor performance compared to sham-injected animals. A safety study in immunodeficient mice showed that intrathecal transplantation of hES-AS is safe. Transplanted hES-AS attached to the meninges along the neuroaxis and survived for the entire duration of the study without formation of tumors or teratomas. Cell-injected mice gained similar body weight to the sham-injected group and did not exhibit clinical signs that could be related to the treatment. No differences from the vehicle control were observed in hematological parameters or blood chemistry. CONCLUSION: Our findings demonstrate the safety and potential therapeutic benefits of intrathecal injection of hES-AS for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Human Embryonic Stem Cells/metabolism , Injections, Spinal/methods , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Humans , Mice , Rats , Superoxide Dismutase-1/metabolism
5.
Stem Cell Reports ; 10(5): 1439-1441, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29742390

ABSTRACT

Following Jamie Thomson's lecture on primate embryonic stem cells (ESCs) at a meeting I had organized in March 1997, in Israel, to celebrate receipt of the Wolf Prize in Agriculture to my colleague and friend Neal First, frozen human embryos donated for research in Israel were shipped to Wisconsin. The five hESC lines (H1, H7, H9, H13, and H14) were established by early 1998 and transferred to my laboratory just before publication of their existence in Science, on November 6, 1998. The distribution of the cells from my institute to several laboratories, as early as 1999, enhanced the development of hESC research worldwide. My personal perspective regarding the scientific and political events surrounding this story are presented.


Subject(s)
Cell Separation/methods , Human Embryonic Stem Cells/cytology , Cell Line , Embryo Culture Techniques , Embryo, Mammalian/cytology , Humans , Stem Cell Research
6.
Adv Biochem Eng Biotechnol ; 163: 23-37, 2018.
Article in English | MEDLINE | ID: mdl-29085956

ABSTRACT

Large-scale expansion of pluripotent stem cells (PSC) in a robust, well-defined, and monitored process is essential for production of cell-based therapeutic products. The transition from laboratory-scale protocols to industrial-scale production is one of the first milestones to be achieved in order to use both human embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) as the starting material for cellular products. The methods to be developed require adjustment of the culture platforms, optimization of culture parameters, and adaptation of downstream procedures. Optimization of expansion protocols and their scalability has become much easier with the design of bioreactor systems that enable continuous monitoring of culture parameters, continuous media change, and support software for automated control. This chapter highlights the common properties that are required for production of scalable, reproducible, homogeneous, and clinically suitable cell therapy products. We describe the available platforms for large-scale expansion of PSCs and parameters that should be considered when optimizing the expansion protocols in a scalable bioreactor. All the above are detailed in the light of the requirements and challenges of bringing a cell-based therapeutic product to the clinic and ultimately to the market. We discuss some considerations that should be taken into account, such as cost-effectiveness, good manufacturing practice, and regulatory guidelines. Graphical Abstract.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism
7.
J Cell Mol Med ; 19(8): 2006-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26153920

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited arrhythmia characterized by syncope and sudden death occurring during exercise or acute emotion. CPVT is caused by abnormal intracellular Ca(2+) handling resulting from mutations in the RyR2 or CASQ2 genes. Because CASQ2 and RyR2 are involved in different aspects of the excitation-contraction coupling process, we hypothesized that these mutations are associated with different functional and intracellular Ca(²+) abnormalities. To test the hypothesis we generated induced Pluripotent Stem Cell-derived cardiomyocytes (iPSC-CM) from CPVT1 and CPVT2 patients carrying the RyR2(R420Q) and CASQ2(D307H) mutations, respectively, and investigated in CPVT1 and CPVT2 iPSC-CM (compared to control): (i) The ultrastructural features; (ii) the effects of isoproterenol, caffeine and ryanodine on the [Ca(2+) ]i transient characteristics. Our major findings were: (i) Ultrastructurally, CASQ2 and RyR2 mutated cardiomyocytes were less developed than control cardiomyocytes. (ii) While in control iPSC-CM isoproterenol caused positive inotropic and lusitropic effects, in the mutated cardiomyocytes isoproterenol was either ineffective, caused arrhythmias, or markedly increased diastolic [Ca(2+) ]i . Importantly, positive inotropic and lusitropic effects were not induced in mutated cardiomyocytes. (iii) The effects of caffeine and ryanodine in mutated cardiomyocytes differed from control cardiomyocytes. Our results show that iPSC-CM are useful for investigating the similarities/differences in the pathophysiological consequences of RyR2 versus CASQ2 mutations underlying CPVT1 and CPVT2 syndromes.


Subject(s)
Calsequestrin/genetics , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/pathology , Base Sequence , Caffeine/pharmacology , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Genotyping Techniques , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Isoproterenol/pharmacology , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure
8.
Hepatology ; 62(1): 265-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808545

ABSTRACT

UNLABELLED: The liver is the main organ responsible for the modification, clearance, and transformational toxicity of most xenobiotics owing to its abundance in cytochrome P450 (CYP450) enzymes. However, the scarcity and variability of primary hepatocytes currently limits their utility. Human pluripotent stem cells (hPSCs) represent an excellent source of differentiated hepatocytes; however, current protocols still produce fetal-like hepatocytes with limited mature function. Interestingly, fetal hepatocytes acquire mature CYP450 expression only postpartum, suggesting that nutritional cues may drive hepatic maturation. We show that vitamin K2 and lithocholic acid, a by-product of intestinal flora, activate pregnane X receptor (PXR) and subsequent CYP3A4 and CYP2C9 expression in hPSC-derived and isolated fetal hepatocytes. Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Flow cytometry showed that over 83% of cells were albumin and hepatocyte nuclear factor 4 alpha (HNF4α) positive, permitting high-content screening in a 96-well plate format. Analysis of 12 compounds showed an R(2) correlation of 0.94 between TC50 values obtained in stem cell-derived hepatocytes and primary cells, compared to 0.62 for HepG2 cells. Finally, stem cell-derived hepatocytes demonstrate all toxicological endpoints examined, including steatosis, apoptosis, and cholestasis, when exposed to nine known hepatotoxins. CONCLUSION: Our work provides fresh insights into liver development, suggesting that microbial-derived cues may drive the maturation of CYP450 enzymes postpartum. Addition of these cues results in the first functional, inducible, hPSC-derived hepatocyte for predictive toxicology.


Subject(s)
Cell Culture Techniques , Hepatocytes/cytology , Lithocholic Acid/pharmacology , Pluripotent Stem Cells/drug effects , Vitamin K 2/pharmacology , Cell Differentiation , Cells, Cultured , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Embryonic Stem Cells/drug effects , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Pregnane X Receptor , Receptors, Steroid/metabolism , Sequence Analysis, RNA , Toxicity Tests, Acute , Vitamin K 2/analogs & derivatives
9.
Cell Metab ; 21(3): 392-402, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25738455

ABSTRACT

Loss of pluripotency is a gradual event whose initiating factors are largely unknown. Here we report the earliest metabolic changes induced during the first hours of differentiation. High-resolution NMR identified 44 metabolites and a distinct metabolic transition occurring during early differentiation. Metabolic and transcriptional analyses showed that pluripotent cells produced acetyl-CoA through glycolysis and rapidly lost this function during differentiation. Importantly, modulation of glycolysis blocked histone deacetylation and differentiation in human and mouse embryonic stem cells. Acetate, a precursor of acetyl-CoA, delayed differentiation and blocked early histone deacetylation in a dose-dependent manner. Inhibitors upstream of acetyl-CoA caused differentiation of pluripotent cells, while those downstream delayed differentiation. Our results show a metabolic switch causing a loss of histone acetylation and pluripotent state during the first hours of differentiation. Our data highlight the important role metabolism plays in pluripotency and suggest that a glycolytic switch controlling histone acetylation can release stem cells from pluripotency.


Subject(s)
Acetyl Coenzyme A/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Glycolysis/physiology , Histones/metabolism , Acetyl Coenzyme A/genetics , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Glycolysis/genetics , Histones/genetics , Humans , Mice , Transcription, Genetic/genetics , Transcription, Genetic/physiology
10.
J Tissue Eng Regen Med ; 9(9): 977-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-23365073

ABSTRACT

Vascularization of injured tissues or artificial grafts is a major challenge in tissue engineering, stimulating a continued search for alternative sources for vasculogenic cells and the development of therapeutic strategies. Human pluripotent stem cells (hPSCs), either embryonic or induced, offer a plentiful platform for the derivation of large numbers of vasculogenic cells, as required for clinical transplantations. Various protocols for generation of vasculogenic smooth muscle cells (SMCs) from hPSCs have been described with considerably different SMC derivatives. In addition, we recently identified hPSC-derived pericytes, which are similar to their physiological counterparts, exhibiting unique features of blood vessel-residing perivascular cells, as well as multipotent mesenchymal precursors with therapeutic angiogenic potential. In this review we refer to methodologies for the development of a variety of perivascular cells from hPSCs with respect to developmental induction, differentiation capabilities, potency and their dual function as mesenchymal precursors. The therapeutic effect of hPSC-derived perivascular cells in experimental models of tissue engineering and regenerative medicine are described and compared to those of their native physiological counterparts.


Subject(s)
Blood Vessels/cytology , Cell Transplantation , Pluripotent Stem Cells/cytology , Animals , Bone Regeneration , Chondrogenesis , Humans , Muscular Dystrophy, Animal/therapy , Myocardial Infarction/therapy , Neovascularization, Physiologic , Skin/cytology
11.
Can J Cardiol ; 30(11): 1279-87, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25442431

ABSTRACT

Human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) have the capacity to differentiate into any specialized cell type, including cardiomyocytes. Therefore, hESC-derived and hiPSC-derived cardiomyocytes (hESC-CMs and hiPSC-CMs, respectively) offer great potential for cardiac regenerative medicine. Unlike some organs, the heart has a limited ability to regenerate, and dysfunction resulting from significant cardiomyocyte loss under pathophysiological conditions, such as myocardial infarction (MI), can lead to heart failure. Unfortunately, for patients with end-stage heart failure, heart transplantation remains the main alternative, and it is insufficient, mainly because of the limited availability of donor organs. Although left ventricular assist devices are progressively entering clinical practice as a bridge to transplantation and even as an optional therapy, cell replacement therapy presents a plausible alternative to donor organ transplantation. During the past decade, multiple candidate cells were proposed for cardiac regeneration, and their mechanisms of action in the myocardium have been explored. The purpose of this article is to critically review the comprehensive research involving the use of hESCs and hiPSCs in MI models and to discuss current controversies, unresolved issues, challenges, and future directions.


Subject(s)
Embryonic Stem Cells/transplantation , Heart Diseases/therapy , Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/pathology , Stem Cell Transplantation/methods , Cell Differentiation , Heart Diseases/pathology , Humans
12.
Tissue Eng Part A ; 20(19-20): 2756-67, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25185111

ABSTRACT

Extracellular matrix (ECM) has been utilized as a biological scaffold for tissue engineering applications in a variety of body systems, due to its bioactivity and biocompatibility. In the current study we developed a modified protocol for the efficient and reproducible derivation of mesenchymal progenitor cells (MPCs) from human embryonic stem cells as well as human induced pluripotent stem cells (hiPSCs) originating from hair follicle keratinocytes (HFKTs). ECM was produced from these MPCs and characterized in comparison to adipose mesenchymal stem cell ECM, demonstrating robust ECM generation by the excised HFKT-iPSC-MPCs. Exploiting the advantages of electrospinning we generated two types of electrospun biodegradable nanofiber layers (NFLs), fabricated from polycaprolactone (PCL) and poly(lactic-co-glycolic acid) (PLGA), which provide mechanical support for cell seeding and ECM generation. Elucidating the optimized decellularization treatment we were able to generate an available "off-the-shelf" implantable product (NFL-ECM). Using rat subcutaneous transplantation model we demonstrate that this stem-cell-derived construct is biocompatible and biodegradable and holds great potential for tissue regeneration applications.


Subject(s)
Extracellular Matrix/chemistry , Induced Pluripotent Stem Cells/metabolism , Lactic Acid/chemistry , Nanofibers/chemistry , Polyesters/chemistry , Polyglycolic Acid/chemistry , Regenerative Medicine , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Keratinocytes/cytology , Keratinocytes/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Polylactic Acid-Polyglycolic Acid Copolymer , Rats
13.
Stem Cells ; 32(12): 3137-49, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25186014

ABSTRACT

Human embryonic stem cells (hESCs) represent a potential source of transplantable cells for regenerative medicine, but development of teratoma even in syngenic recipients represents a critical obstacle to safe stem cell-based therapies. We hypothesized that hESCs escape the immune surveillance by regulating the environmental immune system. Using cocultures of hESCs with allogenic peripheral blood mononuclear cells, we demonstrated that hESCs prevent proliferation and activation of human CD4+ T lymphocytes, an effect dependent upon monocytes. Altered expression of key signaling molecules responsible for the crosstalk of monocytes with T cells was detected in the presence of hESCs. Analyzing the mechanism of action, we demonstrated that hESCs were able to downregulate intracellular glutathione levels in both monocytes and CD4+ cells by suppressing glutamate cysteine ligase expression and to alter MHCII and CD80 expression in monocytes. These effects were achieved at least partially via TGF-beta signaling, and both monocyte phenotype and GCLC expression were affected by Caspase-3 proteolytic activity. Altogether, our results demonstrate a novel immune-suppressive mechanism used by hESCs.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/cytology , Human Embryonic Stem Cells/metabolism , Lymphocyte Activation/immunology , Signal Transduction , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism , Antigens, CD/immunology , Cell Differentiation/immunology , Coculture Techniques/methods , Humans , Monocytes/cytology , Signal Transduction/physiology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology
14.
Stem Cells Transl Med ; 3(10): 1169-81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25205843

ABSTRACT

Isolated microvessel-residing pericytes and pericytes from human pluripotent stem cells (hPSCs) exhibit mesenchymal stem cell-like characteristics and therapeutic properties. Despite growing interest in pericyte-based stem cell therapy, their immunogenicity and immunomodulatory effects on nonactivated T cells are still poorly defined, in particular those of vasculogenic hPSC pericytes. We found that tissue-embedded and unstimulated cultured hPSC- or tissue-derived pericytes constitutively expressed major histocompatibility complex (MHC) class I and the inhibitory programmed cell death-ligand 1/2 (PD-L1/2) molecules but not MHC class II or CD80/CD86 costimulatory molecules. Pretreatment with inflammatory mediators failed to induce an antigen-presenting cell-like phenotype in stimulated pericytes. CD146+ pericytes from hPSCs did not induce activation and proliferation of allogeneic resting T cells independent of interferon (IFN)-γ prestimulation, similarly to pericytes from human brain or placenta. Instead, pericytes mediated a significant increase in the frequency of allogeneic CD25highFoxP3+ regulatory T cells when cocultured with nonactivated peripheral blood T cells. Furthermore, when peripheral blood CD25high regulatory T cells (Tregs) were depleted from isolated CD3+ T cells, pericytes preferentially induced de novo formation of CD4+CD25highFoxP3+CD127-, suppressive regulatory T cells. Constitutive expression of PD-L1/2 and secretion of transforming growth factor-ß by hPSC pericytes directly regulated generation of pericyte-induced Tregs. Pericytes cotransplanted into immunodeficient mice with allogeneic CD25- T cells maintained a nonimmunogenic phenotype and mediated the development of functional regulatory T cells. Together, these findings reveal a novel feature of pericyte-mediated immunomodulation distinguished from immunosuppression, shared by native tissue pericytes and hPSC pericytes, and support the notion that pericytes can be applied for allogeneic cell therapy.


Subject(s)
Lymphocyte Activation/immunology , Pericytes/immunology , Pluripotent Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Humans , Mice , Mice, Inbred NOD , Mice, SCID
15.
Heart Rhythm ; 11(10): 1808-1818, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052725

ABSTRACT

BACKGROUND: We previously reported that induced pluripotent stem cell-derived cardiomyocytes manifest beat rate variability (BRV) resembling heart rate variability (HRV) in the human sinoatrial node. We now hypothesized the BRV-HRV continuum originates in pacemaker cells. OBJECTIVE: To investigate whether cellular BRV is a source of HRV dynamics, we hypothesized 3 levels of interaction among different cardiomyocyte entities: (1) single pacemaker cells, (2) networks of electrically coupled pacemaker cells, and (3) the in situ sinoatrial node. METHODS: We measured BRV/HRV properties in single pacemaker cells, induced pluripotent stem cell-derived contracting embryoid bodies (EBs), and electrocardiograms from the same individual. RESULTS: Pronounced BRV/HRV was present at all 3 levels. The coefficient of variance of interbeat intervals and Poincaré plot indices SD1 and SD2 for single cells were 20 times greater than those for EBs (P < .05) and the in situ heart (the latter two were similar; P > .05). We also compared BRV magnitude among single cells, small EBs (~5-10 cells), and larger EBs (>10 cells): BRV indices progressively increased with the decrease in the cell number (P < .05). Disrupting intracellular Ca(2+) handling markedly augmented BRV magnitude, revealing a unique bimodal firing pattern, suggesting that intracellular mechanisms contribute to BRV/HRV and the fractal behavior of heart rhythm. CONCLUSION: The decreased BRV magnitude in transitioning from the single cell to the EB suggests that the HRV of in situ hearts originates from the summation and integration of multiple cell-based oscillators. Hence, complex interactions among multiple pacemaker cells and intracellular Ca(2+) handling determine HRV in humans and cardiomyocyte networks.


Subject(s)
Electrocardiography , Heart Rate/physiology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Sinoatrial Node/physiology , Action Potentials/physiology , Adult , Female , Healthy Volunteers , Humans , Middle Aged
16.
Stem Cells Dev ; 23(5): 443-56, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24192045

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing is a post-transcriptional, site-specific modification process that is catalyzed by Adenosine Deaminase Acting on RNA (ADAR) gene family members. Since ADARs act on double-stranded RNA, most A-to-I editing occurs within repetitive elements, particularly Alu elements, as the result of the inherent property of these sequences to fold and form double strands. ADAR1-mediated A-to-I RNA editing was recently implicated in the regulation of human embryonic stem cells (hESCs). Spontaneous and neuronal differentiation of hESC was shown to result in a decrease in A-to-I editing levels. Knockdown of ADAR1 in hESCs results in an elevation of the expression of differentiation-related genes. In addition, we found that hESCs over-expressing ADAR1 could not be generated. The current study shows that the editing levels of induced pluripotent stem cells (iPSCs) change throughout reprogramming, from a source cell level to a level similar to that of hESCs. Up- or down-regulation of the ADAR1 level in human foreskin fibroblast (HFF) cells before induction of reprogramming results in varied reprogramming efficiencies. Furthermore, HFF-iPSC early clones derived from source cells in which the ADAR1 level was down-regulated lose their iPSC properties shortly after iPSC colony formation and instead exhibit characteristics of cancer cells. Taken together, our results imply a role for ADAR1 in the regulation of pluripotency induction as well as in the maintenance of early iPSC properties.


Subject(s)
Adenosine Deaminase/biosynthesis , Cell Differentiation/genetics , Embryonic Stem Cells , Induced Pluripotent Stem Cells , Adenosine Deaminase/genetics , Fibroblasts , Gene Expression Regulation , Gene Knockdown Techniques , Humans , RNA-Binding Proteins
17.
Proc Natl Acad Sci U S A ; 110(18): E1685-94, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589888

ABSTRACT

Proper expression and function of the cardiac pacemaker is a critical feature of heart physiology. Two main mechanisms have been proposed: (i) the "voltage-clock," where the hyperpolarization-activated funny current If causes diastolic depolarization that triggers action potential cycling; and (ii) the "Ca(2+) clock," where cyclical release of Ca(2+) from Ca(2+) stores depolarizes the membrane during diastole via activation of the Na(+)-Ca(2+) exchanger. Nonetheless, these mechanisms remain controversial. Here, we used human embryonic stem cell-derived cardiomyocytes (hESC-CMs) to study their autonomous beating mechanisms. Combined current- and voltage-clamp recordings from the same cell showed the so-called "voltage and Ca(2+) clock" pacemaker mechanisms to operate in a mutually exclusive fashion in different cell populations, but also to coexist in other cells. Blocking the "voltage or Ca(2+) clock" produced a similar depolarization of the maximal diastolic potential (MDP) that culminated by cessation of action potentials, suggesting that they converge to a common pacemaker component. Using patch-clamp recording, real-time PCR, Western blotting, and immunocytochemistry, we identified a previously unrecognized Ca(2+)-activated intermediate K(+) conductance (IK(Ca), KCa3.1, or SK4) in young and old stage-derived hESC-CMs. IK(Ca) inhibition produced MDP depolarization and pacemaker suppression. By shaping the MDP driving force and exquisitely balancing inward currents during diastolic depolarization, IK(Ca) appears to play a crucial role in human embryonic cardiac automaticity.


Subject(s)
Embryonic Stem Cells/cytology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Sinoatrial Node/cytology , Sinoatrial Node/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Cell Line , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Models, Cardiovascular , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Sinoatrial Node/drug effects , Thiourea/analogs & derivatives , Thiourea/pharmacology
19.
Am J Obstet Gynecol ; 208(3): 213.e1-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23433325

ABSTRACT

OBJECTIVE: Maternal infection or inflammation may induce fetal inflammatory responses associated with fetal injury and cerebral palsy. We sought to assess the inflammation-associated neuroprotective potential of prophylactic N-acetyl-cysteine (NAC). We examined the effect of NAC on prevention of maternal lipopolysaccharide (LPS)-induced neonatal brain injury using magnetic resonance imaging. STUDY DESIGN: Pregnant Sprague Dawley dams (n = 5-8) at embryonic day 18 received intraperitoneal injection of LPS or saline at time 0. Animals were randomized to receive 2 intravenous injections of NAC or saline (time -30 and 120 minutes). Pups were delivered spontaneously and allowed to mature until postnatal day 25. Female offspring were examined by magnetic resonance brain imaging and analyzed using voxel-based analysis after spatial normalization. T2 relaxation time was used to assess white matter injury and diffusion tensor imaging for apparent diffusion coefficient (ADC) to assess white and gray matter injury. RESULTS: Offspring of LPS-treated dams exhibited significantly increased T2 levels and increased ADC levels in white and gray matter (eg, hypothalamus, motor cortex, corpus callosum, thalamus, hippocampus), consistent with diffuse cerebral injury. In contrast, offspring of NAC-treated LPS dams demonstrated similar T2 and ADC levels as control in both white and gray matter. CONCLUSION: Maternal NAC treatment significantly reduced evidence of neonatal brain injury associated with maternal LPS. These studies suggest that maternal NAC therapy may be effective in human deliveries associated with maternal/fetal inflammation.


Subject(s)
Acetylcysteine/pharmacology , Animals, Newborn , Brain Injuries/prevention & control , Brain/drug effects , Infectious Disease Transmission, Vertical , Inflammation/prevention & control , Pregnancy, Animal , Prenatal Exposure Delayed Effects/prevention & control , Animals , Brain/pathology , Brain Injuries/chemically induced , Brain Injuries/pathology , Female , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides , Magnetic Resonance Imaging , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...