Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
bioRxiv ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38979277

ABSTRACT

Inter-cellular transmission of mRNA is being explored in mammalian species using immortal cell lines (1-3). Here, we uncover an inter-cellular mRNA transfer phenomenon that allows for the adaptation and reprogramming of human primed pluripotent stem cells (hPSCs). This process is induced by the direct cell contact-mediated coculture with mouse embryonic stem cells (mESCs) under the condition impermissible for human primed PSC culture. Mouse-derived mRNA contents are transmitted into adapted hPSCs only in the coculture. Transfer-specific mRNA analysis show the enrichment for divergent biological pathways involving transcription/translational machinery and stress-coping mechanisms, wherein such transfer is diminished when direct cell contacts are lost. After 5 days of mESC culture, surface marker analysis, and global gene profiling confirmed that mRNA transfer-prone hPSC efficiently gains a naïve-like state. Furthermore, transfer-specific knockdown experiments targeting mouse-specific transcription factor-coding mRNAs in hPSC show that mouse-derived Tfcp2l1, Tfap2c, and Klf4 are indispensable for human naïve-like conversion. Thus, inter-species mRNA transfer triggers cellular reprogramming in mammalian cells. Our results support that episodic mRNA transfer can occur in cell cooperative and competitive processes(4), which provides a fresh perspective on understanding the roles of mRNA mobility for intra- and inter-species cellular communications.

2.
Nat Commun ; 15(1): 3366, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684678

ABSTRACT

Autologous skin grafting is a standard treatment for skin defects such as burns. No artificial skin substitutes are functionally equivalent to autologous skin grafts. The cultured epidermis lacks the dermis and does not engraft deep wounds. Although reconstituted skin, which consists of cultured epidermal cells on a synthetic dermal substitute, can engraft deep wounds, it requires the wound bed to be well-vascularized and lacks skin appendages. In this study, we successfully generate complete skin grafts with pluripotent stem cell-derived epidermis with appendages on p63 knockout embryos' dermis. Donor pluripotent stem cell-derived keratinocytes encroach the embryos' dermis by eliminating p63 knockout keratinocytes based on cell-extracellular matrix adhesion mediated cell competition. Although the chimeric skin contains allogenic dermis, it is engraftable as long as autologous grafts. Furthermore, we could generate semi-humanized skin segments by human keratinocytes injection into the amnionic cavity of p63 knockout mice embryos. Niche encroachment opens the possibility of human skin graft production in livestock animals.


Subject(s)
Dermis , Keratinocytes , Mice, Knockout , Skin Transplantation , Animals , Skin Transplantation/methods , Keratinocytes/cytology , Keratinocytes/transplantation , Humans , Dermis/cytology , Dermis/transplantation , Mice , Epidermis/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Skin, Artificial , Epidermal Cells/transplantation , Epidermal Cells/cytology , Extracellular Matrix/metabolism , Skin/cytology
4.
Nature ; 626(7998): 357-366, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052228

ABSTRACT

Recently, several studies using cultures of human embryos together with single-cell RNA-seq analyses have revealed differences between humans and mice, necessitating the study of human embryos1-8. Despite the importance of human embryology, ethical and legal restrictions have limited post-implantation-stage studies. Thus, recent efforts have focused on developing in vitro self-organizing models using human stem cells9-17. Here, we report genetic and non-genetic approaches to generate authentic hypoblast cells (naive hPSC-derived hypoblast-like cells (nHyCs))-known to give rise to one of the two extraembryonic tissues essential for embryonic development-from naive human pluripotent stem cells (hPSCs). Our nHyCs spontaneously assemble with naive hPSCs to form a three-dimensional bilaminar structure (bilaminoids) with a pro-amniotic-like cavity. In the presence of additional naive hPSC-derived analogues of the second extraembryonic tissue, the trophectoderm, the efficiency of bilaminoid formation increases from 20% to 40%, and the epiblast within the bilaminoids continues to develop in response to trophectoderm-secreted IL-6. Furthermore, we show that bilaminoids robustly recapitulate the patterning of the anterior-posterior axis and the formation of cells reflecting the pregastrula stage, the emergence of which can be shaped by genetically manipulating the DKK1/OTX2 hypoblast-like domain. We have therefore successfully modelled and identified the mechanisms by which the two extraembryonic tissues efficiently guide the stage-specific growth and progression of the epiblast as it establishes the post-implantation landmarks of human embryogenesis.


Subject(s)
Embryonic Development , Germ Layers , Pluripotent Stem Cells , Humans , Cell Differentiation , Embryo Implantation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Pluripotent Stem Cells/cytology , Interleukin-6/metabolism , Gastrula/cytology , Gastrula/embryology , Amnion/cytology , Amnion/embryology , Amnion/metabolism , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 120(28): e2216564120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37379351

ABSTRACT

Patients with permanent hypoparathyroidism require lifelong replacement therapy to avoid life-threatening complications, The benefits of conventional treatment are limited, however. Transplanting a functional parathyroid gland (PTG) would yield better results. Parathyroid gland cells generated from pluripotent stem cells in vitro to date cannot mimic the physiological responses to extracellular calcium that are essential for calcium homeostasis. We thus hypothesized that blastocyst complementation (BC) could be a better strategy for generating functional PTG cells and compensating loss of parathyroid function. We here describe generation of fully functional PTGs from mouse embryonic stem cells (mESCs) with single-step BC. Using CRISPR-Cas9 knockout of Glial cells missing2 (Gcm2), we efficiently produced aparathyroid embryos for BC. In these embryos, mESCs differentiated into endocrinologically mature PTGs that rescued Gcm2-/- mice from neonatal death. The mESC-derived PTGs responded to extracellular calcium, restoring calcium homeostasis on transplantation into mice surgically rendered hypoparathyroid. We also successfully generated functional interspecies PTGs in Gcm2-/- rat neonates, an accomplishment with potential for future human PTG therapy using xenogeneic animal BC. Our results demonstrate that BC can produce functional endocrine organs and constitute a concept in treatment of hypoparathyroidism.


Subject(s)
Hypoparathyroidism , Parathyroid Glands , Humans , Animals , Mice , Rats , Calcium , Hypoparathyroidism/genetics , Hypoparathyroidism/therapy , Calcium, Dietary , Blastocyst
6.
Front Endocrinol (Lausanne) ; 13: 963282, 2022.
Article in English | MEDLINE | ID: mdl-35992127

ABSTRACT

Pancreas (and islet) transplantation is the only curative treatment for type 1 diabetes patients whose ß-cell functions have been abolished. However, the lack of donor organs has been the major hurdle to save a large number of patients. Therefore, transplantation of animal organs is expected to be an alternative method to solve the serious shortage of donor organs. More recently, a method to generate organs from pluripotent stem cells inside the body of other species has been developed. This interspecies organ generation using blastocyst complementation (BC) is expected to be the next-generation regenerative medicine. Here, we describe the recent advances and future prospects for these two approaches.


Subject(s)
Organogenesis , Pluripotent Stem Cells , Animals , Blastocyst , Organogenesis/physiology , Regenerative Medicine , Transplantation, Heterologous
7.
Sci Rep ; 12(1): 3242, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217706

ABSTRACT

Blastocyst complementation is an intriguing way of generating humanized animals for organ preparation in regenerative medicine and establishing novel models for drug development. Confirming that complemented organs and cells work normally in chimeric animals is critical to demonstrating the feasibility of blastocyst complementation. Here, we generated thymus-complemented chimeric mice, assessed the efficacy of anti-PD-L1 antibody in tumor-bearing chimeric mice, and then investigated T-cell function. Thymus-complemented chimeric mice were generated by injecting C57BL/6 (B6) embryonic stem cells into Foxn1nu/nu morulae or blastocysts. Flow cytometry data showed that the chimeric mouse thymic epithelial cells (TECs) were derived from the B6 cells. T cells appeared outside the thymi. Single-cell RNA-sequencing analysis revealed that the TEC gene-expression profile was comparable to that in B6 mice. Splenic T cells of chimeric mice responded very well to anti-CD3 stimulation in vitro; CD4+ and CD8+ T cells proliferated and produced IFNγ, IL-2, and granzyme B, as in B6 mice. Anti-PD-L1 antibody treatment inhibited MC38 tumor growth in chimeric mice. Moreover, in the chimeras, anti-PD-L1 antibody restored T-cell activation by significantly decreasing PD-1 expression on T cells and increasing IFNγ-producing T cells in the draining lymph nodes and tumors. T cells produced by complemented thymi thus functioned normally in vitro and in vivo. To successfully generate humanized animals by blastocyst complementation, both verification of the function and gene expression profiling of complemented organs/cells in interspecific chimeras will be important in the near future.


Subject(s)
Blastocyst , CD8-Positive T-Lymphocytes , Animals , Blastocyst/metabolism , Chimera/genetics , Embryonic Stem Cells , Mice , Mice, Inbred C57BL
8.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34874452

ABSTRACT

Despite four decades of effort, robust propagation of pluripotent stem cells from livestock animals remains challenging. The requirements for self-renewal are unclear and the relationship of cultured stem cells to pluripotent cells resident in the embryo uncertain. Here, we avoided using feeder cells or serum factors to provide a defined culture microenvironment. We show that the combination of activin A, fibroblast growth factor and the Wnt inhibitor XAV939 (AFX) supports establishment and continuous expansion of pluripotent stem cell lines from porcine, ovine and bovine embryos. Germ layer differentiation was evident in teratomas and readily induced in vitro. Global transcriptome analyses highlighted commonality in transcription factor expression across the three species, while global comparison with porcine embryo stages showed proximity to bilaminar disc epiblast. Clonal genetic manipulation and gene targeting were exemplified in porcine stem cells. We further demonstrated that genetically modified AFX stem cells gave rise to cloned porcine foetuses by nuclear transfer. In summary, for major livestock mammals, pluripotent stem cells related to the formative embryonic disc are reliably established using a common and defined signalling environment. This article has an associated 'The people behind the papers' interview.


Subject(s)
Cell Differentiation , Embryo, Mammalian/metabolism , Germ Layers/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cattle , Embryo, Mammalian/cytology , Germ Layers/cytology , Livestock , Pluripotent Stem Cells/cytology , Sheep , Species Specificity , Swine
9.
Stem Cell Reports ; 15(1): 125-139, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32531191

ABSTRACT

Two genetic diseases, Gorlin syndrome and McCune-Albright syndrome (MAS), show completely opposite symptoms in terms of bone mineral density and hedgehog (Hh) activity. In this study, we utilized human induced pluripotent stem cell (iPSC)-based models of the two diseases to understand the roles of Hh signaling in osteogenesis. Gorlin syndrome-derived iPSCs showed increased osteoblastogenesis and mineralization with Hh signaling activation and upregulation of a set of transcription factors in an osteogenic culture, compared with the isogenic control. MAS-specific iPSCs showed poor mineralization with low Hh signaling activity in the osteogenic culture; impaired osteoblastogenesis was restored to the normal level by treatment with an Hh signaling-activating small molecule. These data suggest that Hh signaling is a key controller for differentiation of osteoblasts from precursors. This study may pave a path to new drug therapies for genetic abnormalities in calcification caused by dysregulation of Hh signaling.


Subject(s)
Hedgehog Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis , Basal Cell Nevus Syndrome/pathology , Cell Culture Techniques , Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Signal Transduction , Transcription Factors/metabolism , Transcriptome/genetics
10.
Regen Ther ; 14: 19-31, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31988991

ABSTRACT

Clinically relevant human induced pluripotent stem cell (hiPSC) derivatives require efficient protocols to differentiate hiPSCs into specific lineages. Here we developed a fully defined xeno-free strategy to direct hiPSCs toward osteoblasts within 21 days. The strategy successfully achieved the osteogenic induction of four independently derived hiPSC lines by a sequential use of combinations of small-molecule inducers. The induction first generated mesodermal cells, which subsequently recapitulated the developmental expression pattern of major osteoblast genes and proteins. Importantly, Col2.3-Cherry hiPSCs subjected to this strategy strongly expressed the cherry fluorescence that has been observed in bone-forming osteoblasts in vivo. Moreover, the protocol combined with a three-dimensional (3D) scaffold was suitable for the generation of a xeno-free 3D osteogenic system. Thus, our strategy offers a platform with significant advantages for bone biology studies and it will also contribute to clinical applications of hiPSCs to skeletal regenerative medicine.

11.
J Bone Miner Metab ; 38(3): 405-411, 2020 May.
Article in English | MEDLINE | ID: mdl-31897746

ABSTRACT

INTRODUCTION: Because aging is a predictor of renal insufficiency in the general population, renal function is a concern in postmenopausal patients undergoing treatment for osteoporosis. Although high serum phosphate concentration is a predictor of renal insufficiency, the effect of selective estrogen receptor modulator (SERM) on renal function and phosphate homeostasis remains to be established. MATERIALS AND METHODS: We administered 20 mg/day bazedoxifene to 48 postmenopausal osteoporotic women who had been taking alfacalcidol for ≥ 6 months, and assessed lumbar spine bone mineral density (LS-BMD), renal function (by calculating estimated glomerular filtration rate using serum cystatin-C levels [eGFRcys] [range 38.0-98.2 mL/min/1.73 m2]), and phosphate homeostasis. RESULTS: LS-BMD was significantly higher 6 months after the initiation of bazedoxifene administration. eGFRcys had increased by 3 months after initiation and was stable until 12 months. Serum phosphate gradually decreased after initiation, reaching statistical significance at 6 months. The changes in serum phosphate were also significant when the maximum tubular reabsorption rate of phosphate was normalized to glomerular filtration rate (TmP/GFR), indicating that bazedoxifene treatment reduces serum phosphate by increasing the urinary excretion of phosphate. The change in eGFRcys after the initiation of bazedoxifene was significantly negatively correlated with the change in serum phosphate, suggesting that a reduction in serum phosphate improves renal function. CONCLUSION: Bazedoxifene improves renal function, possibly by increasing renal phosphate excretion, in postmenopausal osteoporotic women without severe renal insufficiency.


Subject(s)
Indoles/therapeutic use , Kidney/physiopathology , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/urine , Phosphates/urine , Aged , Bone Density/drug effects , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Glomerular Filtration Rate/drug effects , Homeostasis , Humans , Indoles/pharmacology , Kidney/drug effects , Linear Models , Osteoporosis, Postmenopausal/blood , Osteoporosis, Postmenopausal/physiopathology , Parathyroid Hormone/blood , Phosphates/blood
12.
Stem Cell Reports ; 14(1): 21-33, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31883918

ABSTRACT

We have previously established a concept of developing exogenic pancreas in a genetically modified pig fetus with an apancreatic trait, thereby proposing the possibility of in vivo generation of functional human organs in xenogenic large animals. In this study, we aimed to demonstrate a further proof-of-concept of the compensation for disabled organogeneses in pig, including pancreatogenesis, nephrogenesis, hepatogenesis, and vasculogenesis. These dysorganogenetic phenotypes could be efficiently induced via genome editing of the cloned pigs. Induced dysorganogenetic traits could also be compensated by allogenic blastocyst complementation, thereby proving the extended concept of organ regeneration from exogenous pluripotent cells in empty niches during various organogeneses. These results suggest that the feasibility of blastocyst complementation using genome-edited cloned embryos permits experimentation toward the in vivo organ generation in pigs from xenogenic pluripotent cells.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Organogenesis , Animals , Animals, Genetically Modified , Biomarkers , Cell Differentiation/genetics , Cloning, Organism , Fetus , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Homeodomain Proteins , Organogenesis/genetics , Pancreas/embryology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Regeneration , Swine , Trans-Activators/deficiency
13.
Stem Cell Reports ; 13(3): 530-544, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31402337

ABSTRACT

A simple induction protocol to differentiate chondrocytes from pluripotent stem cells (PSCs) using small-molecule compounds is beneficial for cartilage regenerative medicine and mechanistic studies of chondrogenesis. Here, we demonstrate that chondrocytes are robustly induced from human PSCs by simple combination of two compounds, CHIR99021, a glycogen synthase kinase 3 inhibitor, and TTNPB, a retinoic acid receptor (RAR) agonist, under serum- and feeder-free conditions within 5-9 days. An excellent differentiation efficiency and potential to form hyaline cartilaginous tissues in vivo were demonstrated. Comprehensive gene expression and open chromatin analyses at each protocol stage revealed step-by-step differentiation toward chondrocytes. Genome-wide analysis of RAR and ß-catenin association with DNA showed that retinoic acid and Wnt/ß-catenin signaling collaboratively regulated the key marker genes at each differentiation stage. This method provides a promising cell source for regenerative medicine and, as an in vitro model, may facilitate elucidation of the molecular mechanisms underlying chondrocyte differentiation.


Subject(s)
Benzoates/pharmacology , Cell Differentiation/drug effects , Chondrocytes/metabolism , Pluripotent Stem Cells/cytology , Pyridines/pharmacology , Pyrimidines/pharmacology , Retinoids/pharmacology , Animals , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/cytology , Chondrocytes/transplantation , Chondrogenesis , Chromatin/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression , Humans , Mice , Mice, Inbred NOD , Pluripotent Stem Cells/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
14.
Nat Commun ; 10(1): 451, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30723213

ABSTRACT

Regeneration of human kidneys in animal models would help combat the severe shortage of donors in transplantation therapy. Previously, we demonstrated by interspecific blastocyst complementation between mouse and rats, generation of pluripotent stem cell (PSC)-derived functional pancreas, in apancreatic Pdx1 mutant mice. We, however, were unable to obtain rat PSC-derived kidneys in anephric Sall1 mutant mice, likely due to the poor contribution of rat PSCs to the mouse metanephric mesenchyme, a nephron progenitor. Here, conversely, we show that mouse PSCs can efficiently differentiate into the metanephric mesenchyme in rat, allowing the generation of mouse PSC-derived kidney in anephric Sall1 mutant rat. Glomerular epithelium and renal tubules in the kidneys are entirely composed of mouse PSC-derived cells expressing key functional markers. Importantly, the ureter-bladder junction is normally formed. These data provide proof-of-principle for interspecific blastocyst complementation as a viable approach for kidney generation.


Subject(s)
Kidney Failure, Chronic/therapy , Pluripotent Stem Cells/transplantation , Transcription Factors/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kidney/growth & development , Kidney/metabolism , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/physiopathology , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Organogenesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Rats , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transplantation, Homologous
15.
Sci Rep ; 8(1): 15289, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327488

ABSTRACT

To study development of the conceptus in xenogeneic environments, we assessed interspecies chimera formation as well as tetraploid complementation between mouse and rat. Overall contribution of donor PSC-derived cells was lower in interspecies chimeras than in intraspecies chimeras, and high donor chimerism was associated with anomalies or embryonic death. Organ to organ variation in donor chimerism was greater in interspecies chimeras than in intraspecies chimeras, suggesting species-specific affinity differences among interacting molecules necessary for organogenesis. In interspecies tetraploid complementation, embryo development was near normal until the stage of placental formation, after which no embryos survived.


Subject(s)
Complement System Proteins/immunology , Embryonic Development , Organogenesis , Tetraploidy , Transplantation Chimera , Animals , Blastocyst/cytology , Female , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/cytology , Pregnancy , Rats , Rats, Wistar , Species Specificity , Transplantation Chimera/growth & development , Transplantation Chimera/immunology
16.
Stem Cell Reports ; 11(4): 988-997, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30245211

ABSTRACT

In the case of organ transplantation accompanied by vascular anastomosis, major histocompatibility complex mismatched vascular endothelial cells become a target for graft rejection. Production of a rejection-free, transplantable organ, therefore, requires simultaneous generation of vascular endothelial cells within the organ. To generate pluripotent stem cell (PSC)-derived vascular endothelial cells, we performed blastocyst complementation with a vascular endothelial growth factor receptor-2 homozygous mutant blastocyst. This mutation is embryonic lethal at embryonic (E) day 8.5-9.5 due to an early defect in endothelial and hematopoietic cells. The Flk-1 homozygous knockout chimeric mice survived to adulthood for over 1 year without any abnormality, and all vascular endothelial cells and hematopoietic cells were derived from the injected PSCs. This approach could be used in conjunction with other gene knockouts which induce organ deficiency to produce a rejection-free, transplantable organ in which all the organ's cells and vasculature are PSC derived.


Subject(s)
Blastocyst/cytology , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Aging/metabolism , Animals , Blastocyst/metabolism , Chimera , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Pericytes/cytology , Pericytes/metabolism , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1 , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Diabetes ; 67(11): 2268-2279, 2018 11.
Article in English | MEDLINE | ID: mdl-30115653

ABSTRACT

ß-Cell loss and dysfunction play a critical role in the progression of type 1 and type 2 diabetes. Identifying new molecules and/or molecular pathways that improve ß-cell function and/or increase ß-cell mass should significantly contribute to the development of new therapies for diabetes. Using the zebrafish model, we screened 4,640 small molecules to identify modulators of ß-cell function. This in vivo strategy identified 84 stimulators of insulin expression, which simultaneously reduced glucose levels. The insulin promoter activation kinetics for 32 of these stimulators were consistent with a direct mode of action. A subset of insulin stimulators, including the antidiabetic drug pioglitazone, induced the coordinated upregulation of gluconeogenic pck1 expression, suggesting functional response to increased insulin action in peripheral tissues. Notably, Kv1.3 inhibitors increased ß-cell mass in larval zebrafish and stimulated ß-cell function in adult zebrafish and in the streptozotocin-induced hyperglycemic mouse model. In addition, our data indicate that cytoplasmic Kv1.3 regulates ß-cell function. Thus, using whole-organism screening, we have identified new small-molecule modulators of ß-cell function and glucose metabolism.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Animals, Genetically Modified , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Gene Expression Profiling , Insulin/genetics , Insulin-Secreting Cells/drug effects , Mice , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Pioglitazone/pharmacology , Promoter Regions, Genetic , Up-Regulation/drug effects , Zebrafish
18.
Nature ; 550(7676): 393-397, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29019987

ABSTRACT

Mouse embryonic stem cells derived from the epiblast contribute to the somatic lineages and the germline but are excluded from the extra-embryonic tissues that are derived from the trophectoderm and the primitive endoderm upon reintroduction to the blastocyst. Here we report that cultures of expanded potential stem cells can be established from individual eight-cell blastomeres, and by direct conversion of mouse embryonic stem cells and induced pluripotent stem cells. Remarkably, a single expanded potential stem cell can contribute both to the embryo proper and to the trophectoderm lineages in a chimaera assay. Bona fide trophoblast stem cell lines and extra-embryonic endoderm stem cells can be directly derived from expanded potential stem cells in vitro. Molecular analyses of the epigenome and single-cell transcriptome reveal enrichment for blastomere-specific signature and a dynamic DNA methylome in expanded potential stem cells. The generation of mouse expanded potential stem cells highlights the feasibility of establishing expanded potential stem cells for other mammalian species.


Subject(s)
Blastomeres/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Blastocyst/cytology , Blastomeres/metabolism , Cell Lineage , Cells, Cultured , Chimera , Embryo, Mammalian/cytology , Endoderm/cytology , Epigenesis, Genetic , Epigenomics , Female , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Placenta/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pregnancy , Single-Cell Analysis , Transcriptome , Trophoblasts/cytology
19.
Development ; 144(14): 2544-2547, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28720651

ABSTRACT

Interspecies chimeric assays are a valuable tool for investigating the potential of human stem and progenitor cells, as well as their differentiated progeny. This Spotlight article discusses the different factors that affect interspecies chimera generation, such as evolutionary distance, developmental timing, and apoptosis of the transplanted cells, and suggests some possible strategies to address them. A refined approach to generating interspecies chimeras could contribute not only to a better understanding of cellular potential, but also to understanding the nature of xenogeneic barriers and mechanisms of heterochronicity, to modeling human development, and to the creation of human transplantable organs.


Subject(s)
Stem Cell Research , Transplantation Chimera , Animals , Apoptosis , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Species Specificity , Stem Cell Research/ethics
20.
Nature ; 542(7640): 191-196, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28117444

ABSTRACT

Islet transplantation is an established therapy for diabetes. We have previously shown that rat pancreata can be created from rat pluripotent stem cells (PSCs) in mice through interspecies blastocyst complementation. Although they were functional and composed of rat-derived cells, the resulting pancreata were of mouse size, rendering them insufficient for isolating the numbers of islets required to treat diabetes in a rat model. Here, by performing the reverse experiment, injecting mouse PSCs into Pdx-1-deficient rat blastocysts, we generated rat-sized pancreata composed of mouse-PSC-derived cells. Islets subsequently prepared from these mouse-rat chimaeric pancreata were transplanted into mice with streptozotocin-induced diabetes. The transplanted islets successfully normalized and maintained host blood glucose levels for over 370 days in the absence of immunosuppression (excluding the first 5 days after transplant). These data provide proof-of-principle evidence for the therapeutic potential of PSC-derived islets generated by blastocyst complementation in a xenogeneic host.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Heterografts/physiology , Islets of Langerhans Transplantation , Islets of Langerhans/physiology , Organogenesis , Animals , Blastocyst/cytology , Blastocyst/metabolism , Blood Glucose/metabolism , Chimera , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Heterografts/immunology , Homeodomain Proteins , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/immunology , Male , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Rats , Time Factors , Trans-Activators/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...