51
|
Chen S, Du K, Zou C. Current progress in stem cell therapy for type 1 diabetes mellitus. Stem Cell Res Ther 2020; 11:275. [PMID: 32641151 PMCID: PMC7346484 DOI: 10.1186/s13287-020-01793-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is the most common chronic autoimmune disease in young patients and is characterized by the loss of pancreatic β cells; as a result, the body becomes insulin deficient and hyperglycemic. Administration or injection of exogenous insulin cannot mimic the endogenous insulin secreted by a healthy pancreas. Pancreas and islet transplantation have emerged as promising treatments for reconstructing the normal regulation of blood glucose in T1DM patients. However, a critical shortage of pancreases and islets derived from human organ donors, complications associated with transplantations, high cost, and limited procedural availability remain bottlenecks in the widespread application of these strategies. Attempts have been directed to accommodate the increasing population of patients with T1DM. Stem cell therapy holds great potential for curing patients with T1DM. With the advent of research on stem cell therapy for various diseases, breakthroughs in stem cell-based therapy for T1DM have been reported. However, many unsolved issues need to be addressed before stem cell therapy will be clinically feasible for diabetic patients. In this review, we discuss the current research advances in strategies to obtain insulin-producing cells (IPCs) from different precursor cells and in stem cell-based therapies for diabetes.
Collapse
Affiliation(s)
- Shuai Chen
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Kechen Du
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
52
|
Fleischer A, Vallejo-Díez S, Martín-Fernández JM, Sánchez-Gilabert A, Castresana M, Del Pozo A, Esquisabel A, Ávila S, Castrillo JL, Gaínza E, Pedraz JL, Viñas M, Bachiller D. iPSC-Derived Intestinal Organoids from Cystic Fibrosis Patients Acquire CFTR Activity upon TALEN-Mediated Repair of the p.F508del Mutation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:858-870. [PMID: 32373648 PMCID: PMC7195499 DOI: 10.1016/j.omtm.2020.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
Abstract
Cystic fibrosis (CF) is the main genetic cause of death among the Caucasian population. The disease is characterized by abnormal fluid and electrolyte mobility across secretory epithelia. The first manifestations occur within hours of birth (meconium ileus), later extending to other organs, generally affecting the respiratory tract. It is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR encodes a cyclic adenosine monophosphate (cAMP)-dependent, phosphorylation-regulated chloride channel required for transport of chloride and other ions through cell membranes. There are more than 2,000 mutations described in the CFTR gene, but one of them, phenylalanine residue at amino acid position 508 (p.F508del), a recessive allele, is responsible for the vast majority of CF cases worldwide. Here, we present the results of the application of genome-editing techniques to the restoration of CFTR activity in p.F508del patient-derived induced pluripotent stem cells (iPSCs). Gene-edited iPSCs were subsequently used to produce intestinal organoids on which the physiological activity of the restored gene was tested in forskolin-induced swelling tests. The seamless restoration of the p.F508del mutation resulted in normal expression of the mature CFTR glycoprotein, full recovery of CFTR activity, and a normal response of the repaired organoids to treatment with two approved CF therapies: VX-770 and VX-809.
Collapse
Affiliation(s)
- Aarne Fleischer
- Karuna Good Cells Technologies S.L., C/Cercas Bajas 13 Bajo, 01001 Vitoria-Gasteiz, Spain
| | - Sara Vallejo-Díez
- Consejo Superior de Investigaciones Científicas (CSIC/IMEDEA), Miguel Marqués 21, 07190 Esporles, Spain
| | | | | | - Mónica Castresana
- Karuna Good Cells Technologies S.L., C/Cercas Bajas 13 Bajo, 01001 Vitoria-Gasteiz, Spain
| | | | - Amaia Esquisabel
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain.,Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Silvia Ávila
- Genetadi Biotech S.L., Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | | | | | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain.,Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Miguel Viñas
- Laboratory of Molecular Microbiology and Antimicrobials, Department of Pathology and Experimental Therapeutics, University of Barcelona, 08097 Barcelona, Spain
| | - Daniel Bachiller
- Consejo Superior de Investigaciones Científicas (CSIC/IMEDEA), Miguel Marqués 21, 07190 Esporles, Spain
| |
Collapse
|
53
|
Kaneko H, Kaitsuka T, Tomizawa K. Response to Stimulations Inducing Circadian Rhythm in Human Induced Pluripotent Stem Cells. Cells 2020; 9:cells9030620. [PMID: 32143467 PMCID: PMC7140533 DOI: 10.3390/cells9030620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/14/2020] [Accepted: 03/02/2020] [Indexed: 12/23/2022] Open
Abstract
Regenerative medicine and disease modeling are expanding rapidly, through the development of human-induced pluripotent stem cells (hiPSCs). Many exogeneous supplements are often used for the directed differentiation of hiPSCs to specific lineages, such as chemicals and hormones. Some of these are known to synchronize the circadian clock, like forskolin (Frk) and dexamethasone (Dex); however, the response to these stimulations has not been fully elucidated for hiPSCs. In this study, we examined the response of clock genes to synchronizing stimulation, and compared it with fully differentiated cells, U2OS, and fibroblasts. The expression of clock genes did not show circadian rhythms in hiPSCs with Frk and Dex, which could be due to the significantly low levels of BMAL1. On the other hand, a circadian-like rhythm of D-box binding protein (DBP) expression was observed in hiPSCs by culturing them in an environment with a simulated body temperature. However, the inhibition of temperature-inducible factors, which are involved in temperature rhythm-induced synchronization, could not repress the expression of such rhythms, while the inhibition of HIF-1α significantly repressed them. In summary, we suggest that clock genes do not respond to the synchronizing agents in hiPSCs; instead, a unique circadian-like rhythm is induced by the temperature rhythm.
Collapse
Affiliation(s)
| | - Taku Kaitsuka
- Correspondence: (T.K.); (K.T.); Tel.: +81-96-373-5051 (T.K.); +81-96-373-5050 (K.T.)
| | - Kazuhito Tomizawa
- Correspondence: (T.K.); (K.T.); Tel.: +81-96-373-5051 (T.K.); +81-96-373-5050 (K.T.)
| |
Collapse
|
54
|
Awan M, Buriak I, Fleck R, Fuller B, Goltsev A, Kerby J, Lowdell M, Mericka P, Petrenko A, Petrenko Y, Rogulska O, Stolzing A, Stacey GN. Dimethyl sulfoxide: a central player since the dawn of cryobiology, is efficacy balanced by toxicity? Regen Med 2020; 15:1463-1491. [PMID: 32342730 DOI: 10.2217/rme-2019-0145] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dimethyl sulfoxide (DMSO) is the cryoprotectant of choice for most animal cell systems since the early history of cryopreservation. It has been used for decades in many thousands of cell transplants. These treatments would not have taken place without suitable sources of DMSO that enabled stable and safe storage of bone marrow and blood cells until needed for transfusion. Nevertheless, its effects on cell biology and apparent toxicity in patients have been an ongoing topic of debate, driving the search for less cytotoxic cryoprotectants. This review seeks to place the toxicity of DMSO in context of its effectiveness. It will also consider means of reducing its toxic effects, the alternatives to its use and their readiness for active use in clinical settings.
Collapse
Affiliation(s)
- Maooz Awan
- Institute for Liver & Digestive Health, UCL Division of Medicine, Royal Free Hospital, UCL, London, NW3 2PF, UK
| | - Iryna Buriak
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Roland Fleck
- Centre for Ultrastructural Imaging, Kings College London, London, SE1 1UL, UK
| | - Barry Fuller
- Department of Surgical Biotechnology, UCL Division of Surgery, Royal Free Hospital, UCL, London, NW3 2QG, UK
| | - Anatoliy Goltsev
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Julie Kerby
- Cell & Gene Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Mark Lowdell
- Centre for Cell, Gene & Tissue Therapy, Royal Free London NHS FT & UCL, London, NW3 2PF, UK
| | - Pavel Mericka
- Tissue Bank, University Hospital Hradec Kralové, Czech Republic
| | - Alexander Petrenko
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Yuri Petrenko
- Department of Biomaterials & Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Olena Rogulska
- Institute for Problems of Cryobiology & Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavska 23, 61016, Kharkiv
| | - Alexandra Stolzing
- University of Loughborough, Centre for Biological Engineering, Loughborough University, Holywell Park, Loughborough, UK
| | - Glyn N Stacey
- International Stem Cell Banking Initiative, 2 High Street, Barley, Hertfordshire, SG8 8HZ
- Beijing Stem Cell Bank, Institute of Zoology, Chinese Academy of Sciences, 25–2 Beishuan West, Haidan District, 100190 Beijing, China
- Institute of Stem Cells & Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
55
|
Huang H, Rey-Bedón C, Yarmush ML, Usta OB. Deep-supercooling for extended preservation of adipose-derived stem cells. Cryobiology 2020; 92:67-75. [PMID: 31751557 PMCID: PMC7195234 DOI: 10.1016/j.cryobiol.2019.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/04/2023]
Abstract
Cell preservation is an enabling technology for widespread distribution and applications of mammalian cells. Traditional cryopreservation via slow-freezing or vitrification provides long-term storage but requires cytotoxic cryoprotectants (CPA) and tedious CPA loading/unloading, cooling, and recovering procedures. Hypothermic storage around 0-4 °C is an alternative method but only works for a short period due to its high storage temperatures. Here, we report on the deep-supercooling (DSC) preservation of human adipose-derived stem cells at deep subzero temperatures without freezing for extended storage. Enabled by surface sealing with an immiscible oil phase, cell suspension can be preserved in a liquid state at -13 °C and -16 °C for 7 days with high cell viability, retention of stemness, attachment, and multilineage differentiation capacities. These results demonstrate that DSC is an improved short-term preservation approach to provide off-the-shelf cell sources for booming cell-based medicine and bioengineering.
Collapse
Affiliation(s)
- Haishui Huang
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States
| | - Camilo Rey-Bedón
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, United States.
| | - O Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, United States.
| |
Collapse
|
56
|
van den Brink L, Brandão KO, Yiangou L, Mol MPH, Grandela C, Mummery CL, Verkerk AO, Davis RP. Cryopreservation of human pluripotent stem cell-derived cardiomyocytes is not detrimental to their molecular and functional properties. Stem Cell Res 2020; 43:101698. [PMID: 31945612 PMCID: PMC7611364 DOI: 10.1016/j.scr.2019.101698] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a powerful platform for in vitro modelling of cardiac diseases, safety pharmacology and drug screening. All these applications require large quantities of well-characterised and standardised batches of hiPSC-CMs. Cryopreservation of hiPSC-CMs without affecting their biochemical or biophysical phenotype is essential for facilitating this, but ideally requires the cells being unchanged by the freeze-thaw procedure. We therefore compared the in vitro functional and molecular characteristics of fresh and cryopreserved hiPSC-CMs generated from multiple independent hiPSC lines. While the frozen hiPSC-CMs exhibited poorer replating than their freshly-derived counterparts, there was no difference in the proportion of cardiomyocytes retrieved from the mixed population when this was factored in, although for several lines a higher percentage of ventricular-like hiPSC-CMs were recovered following cryopreservation. Furthermore, cryopreserved hiPSC-CMs from one line exhibited longer action potential durations. These results provide evidence that cryopreservation does not compromise the in vitro molecular, physiological and mechanical properties of hiPSC-CMs, though can lead to an enrichment in ventricular myocytes. It also validates this procedure for storing hiPSC-CMs, thereby allowing the same batch of hiPSC-CMs to be used for multiple applications and evaluations.
Collapse
Affiliation(s)
- Lettine van den Brink
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Karina O Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Mervyn P H Mol
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Catarina Grandela
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, 1105 AZ Amsterdam, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
57
|
Zhang Y, Wang H, Stewart S, Jiang B, Ou W, Zhao G, He X. Cold-Responsive Nanoparticle Enables Intracellular Delivery and Rapid Release of Trehalose for Organic-Solvent-Free Cryopreservation. NANO LETTERS 2019; 19:9051-9061. [PMID: 31680526 DOI: 10.1021/acs.nanolett.9b04109] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Conventional cryopreservation of mammalian cells requires the use of toxic organic solvents (e.g., dimethyl sulfoxide) as cryoprotectants. Consequently, the cryopreserved cells must undergo a tedious washing procedure to remove the organic solvents for their further applications in cell-based medicine, and many of the precious cells may be lost or killed during the procedure. Trehalose has been explored as a nontoxic alternative to traditional cryoprotectants. However, mammalian cells do not synthesize trehalose or express trehalose transporters in their membranes, and the lack of an approach for the efficient intracellular delivery of trehalose has been a major hurdle for its use in cell cryopreservation. In this study, a cold-responsive polymer (poly(N-isopropylacrylamide-co-butyl acrylate)) is utilized to synthesize nanoparticles for the encapsulation and intracellular delivery of trehalose. The trehalose-laden nanoparticles can be efficiently taken up by mammalian cells. The nanoparticles quickly and irreversibly disassemble upon cold treatment, enabling the controlled and rapid release of trehalose from the nanoparticles inside cells. The latter is confirmed by an evident increase in cell volume upon cold treatment. This rapid cold-triggered intracellular release of trehalose is crucial to developing a fast protocol to cryopreserve cells using trehalose. Cells with intracellular trehalose delivered using the nanoparticles show comparable postcryopreservation viability compared to that of cells treated with DMSO, eliminating the need for the tedious and cell-damaging washing procedure required for using the DMSO-cryopreserved cells in vivo. This cold-responsive nanoparticle may greatly facilitate the use of trehalose as a nontoxic cryoprotectant for banking cells and tissues to meet their high demand by modern cell-based medicine.
Collapse
Affiliation(s)
- Yuntian Zhang
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei , Anhui 230027 , China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | | | | | | | - Gang Zhao
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei , Anhui 230027 , China
| | - Xiaoming He
- Marlene and Stewart Greenebaum Comprehensive Cancer Center , University of Maryland , Baltimore , Maryland 21201 , United States
| |
Collapse
|
58
|
Li Q, Huang Q. Single-cell qPCR demonstrates that Repsox treatment changes cell fate from endoderm to neuroectoderm and disrupts epithelial-mesenchymal transition. PLoS One 2019; 14:e0223724. [PMID: 31600351 PMCID: PMC6786562 DOI: 10.1371/journal.pone.0223724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 09/26/2019] [Indexed: 01/22/2023] Open
Abstract
A definitive endodermal cell lineage is a prerequisite for the efficient generation of mature endoderm derivatives that give rise to organs, such as the pancreas and liver. We previously reported that the induction of mesenchymal definitive endoderm cells depends on autocrine TGF-β signaling and that pharmacological blockage of TGF-β signaling by Repsox disrupts endoderm specification. The definitive endoderm arises from a primitive streak, which depends largely on TGF-β signaling. If the TGF-β pathway is blocked by Repsox, cell fate after the primitive streak induction is so-far unknown. We report here, that an induced primitive streak cell-population contained many T/SOX2 co-expressing cells, and subsequent inhibition of TGF-β signaling by Repsox promoted neuroectodermal cell fate, which was characterized using single-cell qPCR analysis and immunostaining. The process of epithelial-to-mesenchymal transition, which is inherent to the process of definitive endoderm differentiation, was also disrupted upon Repsox treatment. Our findings may provide a new approach to produce neural progenitors.
Collapse
Affiliation(s)
- Qiuhong Li
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
- South China Institute of Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- * E-mail:
| | - Qingsong Huang
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
59
|
Jang J, Han D, Golkaram M, Audouard M, Liu G, Bridges D, Hellander S, Chialastri A, Dey SS, Petzold LR, Kosik KS. Control over single-cell distribution of G1 lengths by WNT governs pluripotency. PLoS Biol 2019; 17:e3000453. [PMID: 31557150 PMCID: PMC6782112 DOI: 10.1371/journal.pbio.3000453] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/08/2019] [Accepted: 09/04/2019] [Indexed: 01/10/2023] Open
Abstract
The link between single-cell variation and population-level fate choices lacks a mechanistic explanation despite extensive observations of gene expression and epigenetic variation among individual cells. Here, we found that single human embryonic stem cells (hESCs) have different and biased differentiation potentials toward either neuroectoderm or mesendoderm depending on their G1 lengths before the onset of differentiation. Single-cell variation in G1 length operates in a dynamic equilibrium that establishes a G1 length probability distribution for a population of hESCs and predicts differentiation outcome toward neuroectoderm or mesendoderm lineages. Although sister stem cells generally share G1 lengths, a variable proportion of cells have asymmetric G1 lengths, which maintains the population dispersion. Environmental Wingless-INT (WNT) levels can control the G1 length distribution, apparently as a means of priming the fate of hESC populations once they undergo differentiation. As a downstream mechanism, global 5-hydroxymethylcytosine levels are regulated by G1 length and thereby link G1 length to differentiation outcomes of hESCs. Overall, our findings suggest that intrapopulation heterogeneity in G1 length underlies the pluripotent differentiation potential of stem cell populations. The link between single-cell variation and population-level fate choices lacks a mechanistic explanation. This study finds that the duration of the G1 cell cycle phase in stem cells varies within the population, giving rise to a probability distribution of G1 length that is responsive to Wnt signalling and that predicts cells’ differentiation potential upon exit from pluripotency.
Collapse
Affiliation(s)
- Jiwon Jang
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dasol Han
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Mahdi Golkaram
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Morgane Audouard
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Guojing Liu
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Daniel Bridges
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Physics, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Stefan Hellander
- Department of Computer Science, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Alex Chialastri
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Siddharth S. Dey
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Linda R. Petzold
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Computer Science, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Kenneth S. Kosik
- Department of Molecular, Cellular, and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
- * E-mail:
| |
Collapse
|
60
|
Wang Q, Donelan W, Ye H, Jin Y, Lin Y, Wu X, Wang Y, Xi Y. Real-time observation of pancreatic beta cell differentiation from human induced pluripotent stem cells. Am J Transl Res 2019; 11:3490-3504. [PMID: 31312361 PMCID: PMC6614661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/01/2019] [Indexed: 06/10/2023]
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) into functional insulin-producing cells (IPCs) holds great promise for cell therapy for diabetic patients. Despite recent advances in developing beta cell differentiation protocols, it is becoming clear that the hPSC-derived beta-like cells are functionally immature, and the efficiencies of differentiation can be variable depending on the hPSC lines used. Therefore, advanced methodologies are highly desirable for the development and refinement of beta cell differentiation protocols from hPSCs. In this report, we first derived and validated a Pdx1-mRFP/insulin-hrGFP dual-reporter cell line from MRC5-iPSCs. Then, using this dual-reporter cell line, we developed and optimized an in vitro beta cell differentiation protocol through real-time monitoring expression of Pdx1 and insulin. We demonstrated that DNA demethylation could increase the efficiency of beta cell differentiation. Furthermore, three-dimensional induction not only significantly increased the efficiency of pancreatic progenitor specification and the yield of IPCs, but also produced more mature IPCs. The current study indicates that this dual-reporter cell line is of great value for developing and optimizing the beta cell differentiation protocols. It will facilitate the development of novel protocols for generating IPCs from hPSCs and the investigation of beta cell differentiation mechanisms.
Collapse
Affiliation(s)
- Qiwei Wang
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| | - William Donelan
- Department of Urology, University of FloridaGainesville, Florida 32610, USA
| | - Huahu Ye
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| | - Yulan Jin
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100006, P. R. China
| | - Yanli Lin
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| | - Xiaojie Wu
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| | - Youliang Wang
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| | - Yongyi Xi
- Cell Engineering Laboratory, Beijing Institute of Biotechnology20 Dongda Street, Fengtai District, Beijing 100071, P. R. China
| |
Collapse
|
61
|
Shcherbina A, Li J, Narayanan C, Greenleaf W, Kundaje A, Chetty S. Brief Report: Cell Cycle Dynamics of Human Pluripotent Stem Cells Primed for Differentiation. Stem Cells 2019; 37:1151-1157. [PMID: 31135093 PMCID: PMC6711778 DOI: 10.1002/stem.3041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/26/2019] [Accepted: 05/14/2019] [Indexed: 01/09/2023]
Abstract
Understanding the molecular properties of the cell cycle of human pluripotent stem cells (hPSCs) is critical for effectively promoting differentiation. Here, we use the Fluorescence Ubiquitin Cell Cycle Indicator system adapted into hPSCs and perform RNA sequencing on cell cycle sorted hPSCs primed and unprimed for differentiation. Gene expression patterns of signaling factors and developmental regulators change in a cell cycle‐specific manner in cells primed for differentiation without altering genes associated with pluripotency. Furthermore, we identify an important role for PI3K signaling in regulating the early transitory states of hPSCs toward differentiation. stem cells2019;37:1151–1157
Collapse
Affiliation(s)
- Anna Shcherbina
- Department of Biomedical Informatics, Stanford University, Stanford, California, USA
| | - Jingling Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Cyndhavi Narayanan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - William Greenleaf
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, California, USA.,Department of Computer Science, Stanford University, Stanford, California, USA
| | - Sundari Chetty
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
62
|
Markouli C, Couvreu De Deckersberg E, Regin M, Nguyen HT, Zambelli F, Keller A, Dziedzicka D, De Kock J, Tilleman L, Van Nieuwerburgh F, Franceschini L, Sermon K, Geens M, Spits C. Gain of 20q11.21 in Human Pluripotent Stem Cells Impairs TGF-β-Dependent Neuroectodermal Commitment. Stem Cell Reports 2019; 13:163-176. [PMID: 31178415 PMCID: PMC6627003 DOI: 10.1016/j.stemcr.2019.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
Abstract
Gain of 20q11.21 is one of the most common recurrent genomic aberrations in human pluripotent stem cells. Although it is known that overexpression of the antiapoptotic gene Bcl-xL confers a survival advantage to the abnormal cells, their differentiation capacity has not been fully investigated. RNA sequencing of mutant and control hESC lines, and a line transgenically overexpressing Bcl-xL, shows that overexpression of Bcl-xL is sufficient to cause most transcriptional changes induced by the gain of 20q11.21. Moreover, the differentially expressed genes in mutant and Bcl-xL overexpressing lines are enriched for genes involved in TGF-β- and SMAD-mediated signaling, and neuron differentiation. Finally, we show that this altered signaling has a dramatic negative effect on neuroectodermal differentiation, while the cells maintain their ability to differentiate to mesendoderm derivatives. These findings stress the importance of thorough genetic testing of the lines before their use in research or the clinic. Bcl-xL overexpression drives the transcriptomic profile of 20q11.21 mutant lines 20q11.21 mutant lines downregulate CHCHD2, a known TGF-β pathway modulator Mutant lines differentially express genes involved in TGF-β and SMAD signaling Mutant lines show impaired ectoderm commitment due to TGF-β signaling deregulation
Collapse
Affiliation(s)
- C Markouli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - E Couvreu De Deckersberg
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - M Regin
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - H T Nguyen
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, K7/25 Quang Trung, Danang 550000, Vietnam
| | - F Zambelli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium; Clínica EUGIN, Travessera de les Corts 322, 08029 Barcelona, Spain
| | - A Keller
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - D Dziedzicka
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - J De Kock
- Department of In Vitro Toxicology & Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - L Tilleman
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - F Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - L Franceschini
- Laboratory of Molecular & Cellular Therapy, Department of Immunology - Physiology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - K Sermon
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - M Geens
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - C Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
63
|
New Technologies To Enhance In Vivo Reprogramming for Regenerative Medicine. Trends Biotechnol 2019; 37:604-617. [DOI: 10.1016/j.tibtech.2018.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022]
|
64
|
Akiyama Y, Shinose M, Watanabe H, Yamada S, Kanda Y. Cryoprotectant-free cryopreservation of mammalian cells by superflash freezing. Proc Natl Acad Sci U S A 2019; 116:7738-7743. [PMID: 30936320 PMCID: PMC6475437 DOI: 10.1073/pnas.1808645116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cryopreservation is widely used to maintain backups of cells as it enables the semipermanent storage of cells. During the freezing process, ice crystals that are generated inside and outside the cells can lethally damage the cells. All conventional cryopreservation methods use at least one cryoprotective agent (CPA) to render water inside and outside the cells vitreous or nanocrystallized (near-vitrification) without forming damaging ice crystals. However, CPAs should ideally be avoided due to their cytotoxicity and potential side effects on the cellular state. Herein, we demonstrate the CPA-free cryopreservation of mammalian cells by ultrarapid cooling using inkjet cell printing, which we named superflash freezing (SFF). The SFF cooling rate, which was estimated by a heat-transfer stimulation, is sufficient to nearly vitrify the cells. The experimental results of Raman spectroscopy measurements, and observations with an ultrahigh-speed video camera support the near-vitrification of the droplets under these conditions. Initially, the practical utility of SFF was demonstrated on mouse fibroblast 3T3 cells, and the results were comparable to conventional CPA-assisted methods. Then, the general viability of this method was confirmed on mouse myoblast C2C12 cells and rat primary mesenchymal stem cells. In their entirety, the thus-obtained results unequivocally demonstrate that CPA-free cell cryopreservation is possible by SFF. Such a CPA-free cryopreservation method should be ideally suited for most cells and circumvent the problems typically associated with the addition of CPAs.
Collapse
Affiliation(s)
- Yoshitake Akiyama
- Department of Mechanical Engineering and Robotics, Faculty of Textile Science and Technology, Shinshu University, 3-15-1, Tokida, Ueda, Nagano 386-8567, Japan;
| | - Masato Shinose
- Department of Mechanical Engineering and Robotics, Faculty of Textile Science and Technology, Shinshu University, 3-15-1, Tokida, Ueda, Nagano 386-8567, Japan
| | - Hiroki Watanabe
- Department of Mechanical Engineering and Robotics, Faculty of Textile Science and Technology, Shinshu University, 3-15-1, Tokida, Ueda, Nagano 386-8567, Japan
| | - Shigeru Yamada
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| |
Collapse
|
65
|
Dent MAR, Aranda-Anzaldo A. Lessons we can learn from neurons to make cancer cells quiescent. J Neurosci Res 2019; 97:1141-1152. [PMID: 30985022 DOI: 10.1002/jnr.24428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/20/2019] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
Cancer is a major concern for contemporary societies. However, the incidence of cancer is unevenly distributed among tissues and cell types. In particular, the evidence indicates that neurons are absolutely resistant to cancer and this is commonly explained on the basis of the known postmitotic state of neurons. The dominant paradigm on cancer understands this problem as a disease caused by mutations in cellular genes that result in unrestrained cell proliferation and eventually in tissue invasion and metastasis. However, the evidence also shows that mutations and gross chromosomal anomalies are common in functional neurons that nevertheless do not become neoplastic. This fact suggests that in the real nonexperimental setting mutations per se are not enough for inducing carcinogenesis but also that the postmitotic state of neurons is not genetically controlled or determined, otherwise there should be reports of spontaneously transformed neurons. Here we discuss the evidence that the postmitotic state of neurons has a structural basis on the high stability of their nuclear higher order structure that performs like an absolute tumor suppressor. We also discuss evidence that it is possible to induce a similar structural postmitotic state in nonneural cell types as a practical strategy for stopping or reducing the progression of cancer.
Collapse
Affiliation(s)
- Myrna A R Dent
- Laboratorio de Biología Molecular y Neurociencias, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Armando Aranda-Anzaldo
- Laboratorio de Biología Molecular y Neurociencias, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| |
Collapse
|
66
|
YAP inhibition enhances the differentiation of functional stem cell-derived insulin-producing β cells. Nat Commun 2019; 10:1464. [PMID: 30931946 PMCID: PMC6443737 DOI: 10.1038/s41467-019-09404-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 03/07/2019] [Indexed: 01/27/2023] Open
Abstract
Stem cell-derived insulin-producing beta cells (SC-β) offer an inexhaustible supply of functional β cells for cell replacement therapies and disease modeling for diabetes. While successful directed differentiation protocols for this cell type have been described, the mechanisms controlling its differentiation and function are not fully understood. Here we report that the Hippo pathway controls the proliferation and specification of pancreatic progenitors into the endocrine lineage. Downregulation of YAP, an effector of the pathway, enhances endocrine progenitor differentiation and the generation of SC-β cells with improved insulin secretion. A chemical inhibitor of YAP acts as an inducer of endocrine differentiation and reduces the presence of proliferative progenitor cells. Conversely, sustained activation of YAP results in impaired differentiation, blunted glucose-stimulated insulin secretion, and increased proliferation of SC-β cells. Together these results support a role for YAP in controlling the self-renewal and differentiation balance of pancreatic progenitors and limiting endocrine differentiation in vitro. Pluripotent stem cells can be directed into insulin-producing beta cells in vitro. Here, the authors show that downregulation of YAP, an effector of the Hippo pathway, enhances endocrine progenitor differentiation and the generation of beta-cells with improved insulin secretion.
Collapse
|
67
|
Addressing Variability and Heterogeneity of Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:1-29. [DOI: 10.1007/5584_2019_350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
68
|
Chemically defined and xenogeneic-free differentiation of human pluripotent stem cells into definitive endoderm in 3D culture. Sci Rep 2019; 9:996. [PMID: 30700818 PMCID: PMC6353891 DOI: 10.1038/s41598-018-37650-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into definitive endoderm (DE) represents a key step towards somatic cells of lung, liver and pancreas. For future clinical applications, mass production of differentiated cells at chemically defined conditions and free of xenogeneic substances is envisioned. In this study we adapted our previously published two-dimensional (2D) DE induction protocol to three-dimensional (3D) static suspension culture in the absence of the xenogeneic extracellular matrix Matrigel. Next, fetal calf serum and bovine serum albumin present in the standard medium were replaced by a custom-made and xeno-free B-27. This yielded in a chemically defined and xenogeneic-free 3D culture protocol for differentiation of hPSCs into DE at efficiencies similar to standard 2D conditions. This novel protocol successfully worked with different hPSC lines including hESCs and hiPSCs maintained in two different stem cell media prior to differentiation. DE cells obtained by our novel BSA-free 3D protocol could be further differentiated into PDX1- or NKX6.1-expressing pancreatic progenitor cells. Notably, upon DE differentiation, we also identified a CXCR4+/NCAM+/EpCAMlow cell population with reduced DE marker gene expression. These CXCR4+/NCAM+/EpCAMlow cells emerge as a result of Wnt/beta-catenin hyperactivation via elevated CHIR-99021 concentrations and likely represent misspecified DE.
Collapse
|
69
|
Budash HV, Bilko NM. Embryonic and Induced Pluripotent Stem Cells and Their Differentiation in the Cardiomyocyte Direction in the Presence of Dimethyl Sulfoxide. CYTOL GENET+ 2019. [DOI: 10.3103/s0095452719010055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
70
|
Li J, Narayanan C, Bian J, Sambo D, Brickler T, Zhang W, Chetty S. A transient DMSO treatment increases the differentiation potential of human pluripotent stem cells through the Rb family. PLoS One 2018; 13:e0208110. [PMID: 30540809 PMCID: PMC6291069 DOI: 10.1371/journal.pone.0208110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/12/2018] [Indexed: 01/01/2023] Open
Abstract
The propensity for differentiation varies substantially across human pluripotent stem cell (hPSC) lines, greatly restricting the use of hPSCs for cell replacement therapy or disease modeling. Here, we investigate the underlying mechanisms and demonstrate that activation of the retinoblastoma (Rb) pathway in a transient manner is important for differentiation. In prior work, we demonstrated that pre-treating hPSCs with dimethylsulfoxide (DMSO) before directed differentiation enhanced differentiation potential across all three germ layers. Here, we show that exposure to DMSO improves the efficiency of hPSC differentiation through Rb and by repressing downstream E2F-target genes. While transient inactivation of the Rb family members (including Rb, p107, and p130) suppresses DMSO’s capacity to enhance differentiation across all germ layers, transient expression of a constitutively active (non-phosphorylatable) form of Rb increases the differentiation efficiency similar to DMSO. Inhibition of downstream targets of Rb, such as E2F signaling, also promotes differentiation of hPSCs. More generally, we demonstrate that the duration of Rb activation plays an important role in regulating differentiation capacity.
Collapse
Affiliation(s)
- Jingling Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Cyndhavi Narayanan
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jing Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Danielle Sambo
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Thomas Brickler
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Wancong Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sundari Chetty
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
71
|
Controlling fluid flow to improve cell seeding uniformity. PLoS One 2018; 13:e0207211. [PMID: 30440053 PMCID: PMC6237340 DOI: 10.1371/journal.pone.0207211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/16/2018] [Indexed: 11/19/2022] Open
Abstract
Standard methods for seeding monolayer cell cultures in a multiwell plate or dish do not uniformly distribute cells on the surface. With traditional methods, users find aggregation around the circumference, in the centre, or a combination of the two. This variation is introduced due to the macro scale flow of the cell seeding suspension, and movement of the dish before cells can settle and attach to the surface. Reproducibility between labs, users, and experiments is hampered by this variability in cell seeding. We present a simple method for uniform and user-independent the cell seeding using an easily produced uniform cell seeder (UCS) device. This allows precise control of cell density in a reproducible manner. By containing the cell seeding suspension in a defined volume above the culture surface with the UCS, fluctuations in cell density are minimised. Seeding accuracy, as defined by the actual cell density versus the target seeding density is improved dramatically across users with various levels of expertise. We go on to demonstrate the impact of local variation in cell density on the lineage commitment of human embryonic stem cells (hESCs) towards pancreatic endoderm (PE). Variations in the differentiation profile of cells across a culture well closely mirror variations in cell density introduced by seeding method-with the UCS correcting variations in differentiation efficiency. The UCS device provides a simple and reproducible method for uniform seeding across multiple culture systems.
Collapse
|
72
|
Tunçer S, Gurbanov R, Sheraj I, Solel E, Esenturk O, Banerjee S. Low dose dimethyl sulfoxide driven gross molecular changes have the potential to interfere with various cellular processes. Sci Rep 2018; 8:14828. [PMID: 30287873 PMCID: PMC6172209 DOI: 10.1038/s41598-018-33234-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022] Open
Abstract
Dimethyl sulfoxide (DMSO) is a small molecule with polar, aprotic and amphiphilic properties. It serves as a solvent for many polar and nonpolar molecules and continues to be one of the most used solvents (vehicle) in medical applications and scientific research. To better understand the cellular effects of DMSO within the concentration range commonly used as a vehicle (0.1-1.5%, v/v) for cellular treatments, we applied Attenuated Total Reflectance (ATR) Fourier Transform Infrared (FT-IR) spectroscopy to DMSO treated and untreated epithelial colon cancer cells. Both unsupervised (Principal Component Analysis-PCA) and supervised (Linear Discriminant Analysis-LDA) pattern recognition/modelling algorithms applied to the IR data revealed total segregation and prominent differences between DMSO treated and untreated cells at whole, lipid and nucleic acid regions. Several of these data were supported by other independent techniques. Further IR data analyses of macromolecular profile indicated comprehensive alterations especially in proteins and nucleic acids. Protein secondary structure analysis showed predominance of β-sheet over α-helix in DMSO treated cells. We also observed for the first time, a reduction in nucleic acid level upon DMSO treatment accompanied by the formation of Z-DNA. Molecular docking and binding free energy studies indicated a stabilization of Z-DNA in the presence of DMSO. This alternate DNA form may be related with the specific actions of DMSO on gene expression, differentiation, and epigenetic alterations. Using analytical tools combined with molecular and cellular biology techniques, our data indicate that even at very low concentrations, DMSO induces a number of changes in all macromolecules, which may affect experimental outcomes where DMSO is used as a solvent.
Collapse
Affiliation(s)
- Sinem Tunçer
- Department of Biological Sciences, Orta Dogu Teknik Universitesi (ODTU/METU), Ankara, 06800, Turkey
- Vocational School of Health Services, Department of Medical Laboratory Techniques, Bilecik Şeyh Edebali University, Bilecik, 11230, Turkey
| | - Rafig Gurbanov
- Department of Molecular Biology and Genetics, Bilecik Şeyh Edebali University, Bilecik, 11230, Turkey
| | - Ilir Sheraj
- Department of Biological Sciences, Orta Dogu Teknik Universitesi (ODTU/METU), Ankara, 06800, Turkey
| | - Ege Solel
- Department of Biological Sciences, Orta Dogu Teknik Universitesi (ODTU/METU), Ankara, 06800, Turkey
- Department of Biomedicine, University of Bergen, Postbox 7804, Bergen, N-5020, Norway
| | - Okan Esenturk
- Department of Chemistry, Orta Dogu Teknik Universitesi (ODTU/METU), Ankara, 06800, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi (ODTU/METU), Ankara, 06800, Turkey.
| |
Collapse
|
73
|
Southard SM, Kotipatruni RP, Rust WL. Generation and selection of pluripotent stem cells for robust differentiation to insulin-secreting cells capable of reversing diabetes in rodents. PLoS One 2018; 13:e0203126. [PMID: 30183752 PMCID: PMC6124757 DOI: 10.1371/journal.pone.0203126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 08/14/2018] [Indexed: 01/06/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology enables the creation and selection of pluripotent cells with specific genetic traits. This report describes a pluripotent cell line created specifically to form replacement pancreatic cells as a therapy for insulin-dependent diabetes. Beginning with primary pancreatic tissue acquired through organ donation, cells were isolated, re-programmed using non-integrating vectors and exposed to a four day differentiation protocol to generate definitive endoderm, a developmental precursor to pancreas. The best performing iPSC lines were then subjected to a 12-day basic differentiation protocol to generate endocrine pancreas precursors. The line that most consistently generated highly pure populations was selected for further development. This approach created an iPSC-variant cell line, SR1423, with a genetic profile correlated with preferential differentiation toward endodermal lineage at the loss of mesodermal potential. This report further describes an improved differentiation protocol that, coupled with SR1423, generated populations of greater than 60% insulin-expressing cells that secrete insulin in response to glucose and are capable of reversing diabetes in rodents. Created and banked following cGMP guidelines, SR1423 is a candidate cell line for the production of insulin-producing cells useful for the treatment of diabetes.
Collapse
|
74
|
Zambelli F, Mertens J, Dziedzicka D, Sterckx J, Markouli C, Keller A, Tropel P, Jung L, Viville S, Van de Velde H, Geens M, Seneca S, Sermon K, Spits C. Random Mutagenesis, Clonal Events, and Embryonic or Somatic Origin Determine the mtDNA Variant Type and Load in Human Pluripotent Stem Cells. Stem Cell Reports 2018; 11:102-114. [PMID: 29910126 PMCID: PMC6117474 DOI: 10.1016/j.stemcr.2018.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/10/2018] [Accepted: 05/15/2018] [Indexed: 01/13/2023] Open
Abstract
In this study, we deep-sequenced the mtDNA of human embryonic and induced pluripotent stem cells (hESCs and hiPSCs) and their source cells and found that the majority of variants pre-existed in the cells used to establish the lines. Early-passage hESCs carried few and low-load heteroplasmic variants, similar to those identified in oocytes and inner cell masses. The number and heteroplasmic loads of these variants increased with prolonged cell culture. The study of 120 individual cells of early- and late-passage hESCs revealed a significant diversity in mtDNA heteroplasmic variants at the single-cell level and that the variants that increase during time in culture are always passenger to the appearance of chromosomal abnormalities. We found that early-passage hiPSCs carry much higher loads of mtDNA variants than hESCs, which single-fibroblast sequencing proved pre-existed in the source cells. Finally, we show that these variants are stably transmitted during short-term differentiation.
Collapse
Affiliation(s)
- Filippo Zambelli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium; S.I.S.Me.R. Reproductive Medicine Unit, Via Mazzini 12, Bologna 40100, Italy
| | - Joke Mertens
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | - Dominika Dziedzicka
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | - Johan Sterckx
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, Brussels, Belgium
| | - Christina Markouli
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | - Alexander Keller
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | | | - Laura Jung
- Institut de Parasitologie et Pathologie Tropicale, EA 7292, Fédérationde Médecine Translationelle, Université de Strasbourg, 3 rue Koeberlé, Strasbourg 67000, France
| | - Stephane Viville
- Institut de Parasitologie et Pathologie Tropicale, EA 7292, Fédérationde Médecine Translationelle, Université de Strasbourg, 3 rue Koeberlé, Strasbourg 67000, France; Laboratoire de Diagnostic Génétique, UF3472-génétique de l'infertilité, Hôpitaux Universitaires de Strasbourg, Strasbourg 67000, France
| | - Hilde Van de Velde
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium; Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, Brussels, Belgium
| | - Mieke Geens
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | - Sara Seneca
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium; Centre for Medical Genetics, UZ Brussel, Laarbeeklaan 101, Brussels, Belgium
| | - Karen Sermon
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium
| | - Claudia Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels 1090, Belgium.
| |
Collapse
|
75
|
Stratigopoulos G, De Rosa MC, LeDuc CA, Leibel RL, Doege CA. DMSO increases efficiency of genome editing at two non-coding loci. PLoS One 2018; 13:e0198637. [PMID: 29864154 PMCID: PMC5986138 DOI: 10.1371/journal.pone.0198637] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/22/2018] [Indexed: 11/19/2022] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) has become the tool of choice for genome editing. Despite the fact that it has evolved as a highly efficient means to edit/replace coding sequence, CRISPR/Cas9 efficiency for “clean” editing of non-coding DNA remains low. We set out to introduce a single base-pair substitution in two intronic SNPs at the FTO locus without altering nearby non-coding sequence. Substitution efficiency increased up to 10-fold by treatment of human embryonic stem cells (ESC) with non-toxic levels of DMSO (1%) before CRISPR/Cas9 delivery. Treatment with DMSO did not result in CRISPR/Cas9 off-target effects or compromise the chromosomal stability of the ESC. Twenty-four hour treatment of human ESC with DMSO before CRISPR/Cas9 delivery may prove a simple means to increase editing efficiency of non-coding DNA without incorporation of undesirable mutations.
Collapse
Affiliation(s)
- George Stratigopoulos
- Department of Pediatrics, Columbia University, New York, NY, United States of America
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, United States of America
- * E-mail:
| | - Maria Caterina De Rosa
- Department of Pediatrics, Columbia University, New York, NY, United States of America
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, United States of America
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, United States of America
| | - Charles A. LeDuc
- Department of Pediatrics, Columbia University, New York, NY, United States of America
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, United States of America
- New York Obesity Nutrition Research Center, Columbia University, New York, NY, United States of America
| | - Rudolph L. Leibel
- Department of Pediatrics, Columbia University, New York, NY, United States of America
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, United States of America
- New York Obesity Nutrition Research Center, Columbia University, New York, NY, United States of America
- Institute of Human Nutrition, Columbia University, New York, NY, United States of America
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, United States of America
- Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, United States of America
- New York Obesity Nutrition Research Center, Columbia University, New York, NY, United States of America
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
| |
Collapse
|
76
|
Sambathkumar R, Akkerman R, Dastidar S, Roelandt P, Kumar M, Bajaj M, Mestre Rosa AR, Helsen N, Vanslembrouck V, Kalo E, Khurana S, Laureys J, Gysemans C, Faas MM, de Vos P, Verfaillie CM. Generation of hepatocyte- and endocrine pancreatic-like cells from human induced endodermal progenitor cells. PLoS One 2018; 13:e0197046. [PMID: 29750821 PMCID: PMC5947914 DOI: 10.1371/journal.pone.0197046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 04/25/2018] [Indexed: 01/27/2023] Open
Abstract
Multipotent Adult Progenitor Cells (MAPCs) are one potential stem cell source to generate functional hepatocytes or β-cells. However, human MAPCs have less plasticity than pluripotent stem cells (PSCs), as their ability to generate endodermal cells is not robust. Here we studied the role of 14 transcription factors (TFs) in reprogramming MAPCs to induced endodermal progenitor cells (iENDO cells), defined as cells that can be long-term expanded and differentiated to both hepatocyte- and endocrine pancreatic-like cells. We demonstrated that 14 TF-iENDO cells can be expanded for at least 20 passages, differentiate spontaneously to hepatocyte-, endocrine pancreatic-, gut tube-like cells as well as endodermal tumor formation when grafted in immunodeficient mice. Furthermore, iENDO cells can be differentiated in vitro into hepatocyte- and endocrine pancreatic-like cells. However, the pluripotency TF OCT4, which is not silenced in iENDO cells, may contribute to the incomplete differentiation to mature cells in vitro and to endodermal tumor formation in vivo. Nevertheless, the studies presented here provide evidence that reprogramming of adult stem cells to an endodermal intermediate progenitor, which can be expanded and differentiate to multiple endodermal cell types, might be a valid alternative for the use of PSCs for creation of endodermal cell types.
Collapse
Affiliation(s)
- Rangarajan Sambathkumar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- * E-mail: (CMV); (RS)
| | - Renate Akkerman
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
| | - Sumitava Dastidar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Philip Roelandt
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Manoj Kumar
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Manmohan Bajaj
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Ana Rita Mestre Rosa
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Nicky Helsen
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Veerle Vanslembrouck
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Eric Kalo
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
| | - Satish Khurana
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Jos Laureys
- KU Leuven, Department of Clinical and Experimental Medicine, Clinical and Experimental Endocrinology unit, Leuven, Belgium
| | - Conny Gysemans
- KU Leuven, Department of Clinical and Experimental Medicine, Clinical and Experimental Endocrinology unit, Leuven, Belgium
| | - Marijke M. Faas
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Paul de Vos
- University of Groningen, University Medical Center Groningen (UMCG), Pathology and Medical Biology, Division of Medical Biology, Section Immunoendocrinology, Groningen, The Netherlands
| | - Catherine M. Verfaillie
- KU Leuven, Interdepartmental Stem Cell Institute, Department of Development and Regeneration, Stem Cell Biology and Embryology, Leuven, Belgium
- * E-mail: (CMV); (RS)
| |
Collapse
|
77
|
Liu W, Ren Z, Lu K, Song C, Cheung ECW, Zhou Z, Chen G. The Suppression of Medium Acidosis Improves the Maintenance and Differentiation of Human Pluripotent Stem Cells at High Density in Defined Cell Culture Medium. Int J Biol Sci 2018; 14:485-496. [PMID: 29805300 PMCID: PMC5968841 DOI: 10.7150/ijbs.24681] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/26/2018] [Indexed: 12/17/2022] Open
Abstract
Cell density has profound impacts on the cell culture practices of human pluripotent stem cells. The regulation of cell growth, cell death, pluripotency and differentiation converge at high density, but it is largely unknown how different regulatory mechanisms act at this stage. We use a chemically defined medium to systemically examine cellular activities and the impact of medium components in high-density culture. We show that medium acidosis is the main factor that alters cell cycle, gene expression and cellular metabolism at high cell density. The low medium pH leads to inhibition of glucose consumption, cell cycle arrest, and subsequent cell death. At high cell density, the suppression of medium acidosis with sodium bicarbonate (NaHCO3) significantly increases culture capacity for stem cell survival, derivation, maintenance and differentiation. Our study provides a simple and effective tool to improve stem cell maintenance and applications.
Collapse
Affiliation(s)
- Weiwei Liu
- University of Macau, Faculty of Health Sciences, Taipa, Macau
| | - Zhili Ren
- University of Macau, Faculty of Health Sciences, Taipa, Macau
| | - Kai Lu
- University of Macau, Faculty of Health Sciences, Taipa, Macau
| | - Chengcheng Song
- University of Macau, Faculty of Health Sciences, Taipa, Macau
| | | | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Diagnostic Laboratory Service, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Guokai Chen
- University of Macau, Faculty of Health Sciences, Taipa, Macau
| |
Collapse
|
78
|
Chal J, Al Tanoury Z, Oginuma M, Moncuquet P, Gobert B, Miyanari A, Tassy O, Guevara G, Hubaud A, Bera A, Sumara O, Garnier JM, Kennedy L, Knockaert M, Gayraud-Morel B, Tajbakhsh S, Pourquié O. Recapitulating early development of mouse musculoskeletal precursors of the paraxial mesoderm in vitro. Development 2018; 145:145/6/dev157339. [DOI: 10.1242/dev.157339] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Body skeletal muscles derive from the paraxial mesoderm, which forms in the posterior region of the embryo. Using microarrays, we characterize novel mouse presomitic mesoderm (PSM) markers and show that, unlike the abrupt transcriptome reorganization of the PSM, neural tube differentiation is accompanied by progressive transcriptome changes. The early paraxial mesoderm differentiation stages can be efficiently recapitulated in vitro using mouse and human pluripotent stem cells. While Wnt activation alone can induce posterior PSM markers, acquisition of a committed PSM fate and efficient differentiation into anterior PSM Pax3+ identity further requires BMP inhibition to prevent progenitors from drifting to a lateral plate mesoderm fate. When transplanted into injured adult muscle, these precursors generated large numbers of immature muscle fibers. Furthermore, exposing these mouse PSM-like cells to a brief FGF inhibition step followed by culture in horse serum-containing medium allows efficient recapitulation of the myogenic program to generate myotubes and associated Pax7+ cells. This protocol results in improved in vitro differentiation and maturation of mouse muscle fibers over serum-free protocols and enables the study of myogenic cell fusion and satellite cell differentiation.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Masayuki Oginuma
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Philippe Moncuquet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch Graffenstaden 67400, France
| | - Ayako Miyanari
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Getzabel Guevara
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Alexis Hubaud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Agata Bera
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olga Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Jean-Marie Garnier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Leif Kennedy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Marie Knockaert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Barbara Gayraud-Morel
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| |
Collapse
|
79
|
Keller A, Dziedzicka D, Zambelli F, Markouli C, Sermon K, Spits C, Geens M. Genetic and epigenetic factors which modulate differentiation propensity in human pluripotent stem cells. Hum Reprod Update 2018; 24:162-175. [PMID: 29377992 DOI: 10.1093/humupd/dmx042] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/23/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human pluripotent stem cell (hPSC) lines are known to have a bias in their differentiation. This gives individual cell lines a propensity to preferentially differentiate towards one germ layer or cell type over others. Chromosomal aberrations, mitochondrial mutations, genetic diversity and epigenetic variance are the main drivers of this phenomenon, and can lead to a wide range of phenotypes. OBJECTIVE AND RATIONALE Our aim is to provide a comprehensive overview of the different factors which influence differentiation propensity. Specifically, we sought to highlight known genetic variances and their mechanisms, in addition to more general observations from larger abnormalities. Furthermore, we wanted to provide an up-to-date list of a growing number of predictive indicators which are able to identify differentiation propensity before the initiation of differentiation. As differentiation propensity can lead to difficulties in both research as well as clinical translation, our thorough overview could be a useful tool. SEARCH METHODS Combinations of the following key words were applied as search criteria in the PubMed database: embryonic stem cells, induced pluripotent stem cells, differentiation propensity (also: potential, efficiency, capacity, bias, variability), epigenetics, chromosomal abnormalities, genetic aberrations, X chromosome inactivation, mitochondrial function, mitochondrial metabolism, genetic diversity, reprogramming, predictive marker, residual stem cell, clinic. Only studies in English were included, ranging from 2000 to 2017, with a majority ranging from 2010 to 1017. Further manuscripts were added from cross-references. OUTCOMES Differentiation propensity is affected by a wide variety of (epi)genetic factors. These factors clearly lead to a loss of differentiation capacity, preference towards certain cell types and oftentimes, phenotypes which begin to resemble cancer. Broad changes in (epi)genetics, such as aneuploidies or wide-ranging modifications to the epigenetic landscape tend to lead to extensive, less definite changes in differentiation capacity, whereas more specific abnormalities often have precise ramifications in which certain cell types become more preferential. Furthermore, there appears to be a greater, though often less considered, contribution to differentiation propensity by factors such as mitochondria and inherent genetic diversity. Varied differentiation capacity can also lead to potential consequences in the clinical translation of hPSC, including the occurrence of residual undifferentiated stem cells, and the transplantation of potentially transformed cells. WIDER IMPLICATIONS As hPSC continue to advance towards the clinic, our understanding of them progresses as well. As a result, the challenges faced become more numerous, but also more clear. If the transition to the clinic is to be achieved with a minimum number of potential setbacks, thorough evaluation of the cells will be an absolute necessity. Altered differentiation propensity represents at least one such hurdle, for which researchers and eventually clinicians will need to find solutions. Already, steps are being taken to tackle the issue, though further research will be required to evaluate any long-term risks it poses.
Collapse
Affiliation(s)
- Alexander Keller
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Dominika Dziedzicka
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Filippo Zambelli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Christina Markouli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Karen Sermon
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Claudia Spits
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Mieke Geens
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| |
Collapse
|
80
|
Altan S, Sağsöz H, Oğurtan Z. Topical dimethyl sulfoxide inhibits corneal neovascularization and stimulates corneal repair in rabbits following acid burn. Biotech Histochem 2017; 92:619-636. [DOI: 10.1080/10520295.2017.1371333] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
| | - H Sağsöz
- Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakir
| | - Z Oğurtan
- Department of Surgery, Faculty of Veterinary Medicine, Selçuk University, Konya, Turkey
| |
Collapse
|
81
|
Hemmi JJ, Mishra A, Hornsby PJ. Overcoming barriers to reprogramming and differentiation in nonhuman primate induced pluripotent stem cells. Primate Biol 2017; 4:153-162. [PMID: 32110703 PMCID: PMC7041531 DOI: 10.5194/pb-4-153-2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/17/2017] [Indexed: 11/13/2022] Open
Abstract
Induced pluripotent stem cells (iPS cells) generated by cellular
reprogramming from nonhuman primates (NHPs) are of great significance for
regenerative medicine and for comparative biology. Autologously derived stem
cells would theoretically avoid any risk of rejection due to host–donor
mismatch and may bypass the need for immune suppression post-transplant. In
order for these possibilities to be realized, reprogramming methodologies
that were initially developed mainly for human cells must be translated to
NHPs. NHP studies have typically used pluripotent cells generated from young
animals and thus risk overlooking complications that may arise from
generating iPS cells from donors of other ages. When reprogramming is
extended to a wide range of NHP species, available donors may be middle- or
old-aged. Here we have pursued these questions by generating iPS cells from
donors across the life span of the common marmoset (Callithrix jacchus) and then subjecting them to a directed neural differentiation
protocol. The differentiation potential of different clonal cell lines was
assessed using the quantitative polymerase chain reaction. The results show
that cells derived from older donors often showed less neural marker
induction. These deficits were rescued by a 24 h pretreatment of the cells
with 0.5 % dimethyl sulfoxide. Another NHP that plays a key role in
biological research is the chimpanzee (Pan troglodytes). iPS cells
generated from the chimpanzee can be of great interest in comparative in
vitro studies. We investigated if similar deficits in differentiation
potential might arise in chimpanzee iPS cells reprogrammed using various
technologies. The results show that, while some deficits were observed in iPS
cell clones generated using three different technologies, there was no clear
association with the vector used. These deficits in differentiation were also
prevented by a 24 h pretreatment with 0.5 % dimethyl sulfoxide.
Collapse
Affiliation(s)
- Jacob J Hemmi
- Barshop Institute and Department of Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX 78245, USA
| | - Anuja Mishra
- Barshop Institute and Department of Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX 78245, USA
| | - Peter J Hornsby
- Barshop Institute and Department of Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX 78245, USA
| |
Collapse
|
82
|
Huang H, Zhao G, Zhang Y, Xu J, Toth TL, He X. Predehydration and Ice Seeding in the Presence of Trehalose Enable Cell Cryopreservation. ACS Biomater Sci Eng 2017; 3:1758-1768. [PMID: 28824959 PMCID: PMC5558192 DOI: 10.1021/acsbiomaterials.7b00201] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/11/2017] [Indexed: 12/15/2022]
Abstract
Conventional approaches for cell cryopreservation require the use of toxic membrane-penetrating cryoprotective agents (pCPA), which limits the clinical application of cryopreserved cells. Here, we show intentionally induced ice formation at a high subzero temperature (> -10 °C) during cryopreservation, which is often referred to as ice seeding, could result in significant cell injury in the absence of any pCPA. This issue can be mitigated by predehydrating cells using extracellular trehalose to their minimal volume with minimized osmotically active water before ice seeding. We further observe that ice seeding can minimize the interfacial free energy that drives the devastating ice recrystallization-induced cell injury during warming cryopreserved samples. Indeed, by combining predehydration using extracellular trehalose with ice seeding at high subzero temperatures, high cell viability or recovery is achieved for fibroblasts, adult stem cells, and red blood cells after cryopreservation without using any pCPA. The pCPA-free technology developed in this study may greatly facilitate the long-term storage and ready availability of living cells, tissues, and organs that are of high demand by modern cell-based medicine.
Collapse
Affiliation(s)
- Haishui Huang
- Department
of Biomedical Engineering, The Ohio State
University, 1080 Carmack Road, Columbus, Ohio 43210, United
States
- Department
of Mechanical Engineering, The Ohio State
University, 201 W 19th
Avenue, Columbus, Ohio 43210, United States
| | - Gang Zhao
- Centre
for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230027, China
| | - Yuntian Zhang
- Department
of Biomedical Engineering, The Ohio State
University, 1080 Carmack Road, Columbus, Ohio 43210, United
States
- Centre
for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230027, China
| | - Jiangsheng Xu
- Department
of Biomedical Engineering, The Ohio State
University, 1080 Carmack Road, Columbus, Ohio 43210, United
States
- Davis
Heart and Lung Research Institute, The Ohio
State University, 473
W 12th Avenue, Columbus, Ohio 43210, United
States
- Comprehensive
Cancer Center, The Ohio State University, 460 W 12th Avenue, Columbus, Ohio 43210, United States
| | - Thomas L. Toth
- Vincent Department
of Obstetrics and Gynecology, Vincent Reproductive Medicine and IVF, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, United States
- Department
of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Xiaoming He
- Department
of Biomedical Engineering, The Ohio State
University, 1080 Carmack Road, Columbus, Ohio 43210, United
States
- Davis
Heart and Lung Research Institute, The Ohio
State University, 473
W 12th Avenue, Columbus, Ohio 43210, United
States
- Comprehensive
Cancer Center, The Ohio State University, 460 W 12th Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
83
|
Future Challenges in the Generation of Hepatocyte-Like Cells From Human Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
84
|
Cha JM, Mantalaris A, Jung S, Ji Y, Bang OY, Bae H. Mesoderm Lineage 3D Tissue Constructs Are Produced at Large-Scale in a 3D Stem Cell Bioprocess. Biotechnol J 2017; 12. [PMID: 28731612 DOI: 10.1002/biot.201600748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 06/29/2017] [Indexed: 11/11/2022]
Abstract
Various studies have presented different approaches to direct pluripotent stem cell differentiation such as applying defined sets of exogenous biochemical signals and genetic/epigenetic modifications. Although differentiation to target lineages can be successfully regulated, such conventional methods are often complicated, laborious, and not cost-effective to be employed to the large-scale production of 3D stem cell-based tissue constructs. A 3D-culture platform that could realize the large-scale production of mesoderm lineage tissue constructs from embryonic stem cells (ESCs) is developed. ESCs are cultured using our previously established 3D-bioprocess platform which is amenable to mass-production of 3D ESC-based tissue constructs. Hepatocarcinoma cell line conditioned medium is introduced to the large-scale 3D culture to provide a specific biomolecular microenvironment to mimic in vivo mesoderm formation process. After 5 days of spontaneous differentiation period, the resulting 3D tissue constructs are composed of multipotent mesodermal progenitor cells verified by gene and molecular expression profiles. Subsequently the optimal time points to trigger terminal differentiation towards cardiomyogenesis or osteogenesis from the mesodermal tissue constructs is found. A simple and affordable 3D ESC-bioprocess that can reach the scalable production of mesoderm origin tissues with significantly improved correspondent tissue properties is demonstrated.
Collapse
Affiliation(s)
- Jae Min Cha
- Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Athanasios Mantalaris
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Sunyoung Jung
- Center for Bionics, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yurim Ji
- Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Oh Young Bang
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, 06351, Republic of Korea.,Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Hojae Bae
- KU Convergence Science and Technology Institute, Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, 05029, Republic of Korea
| |
Collapse
|
85
|
Manzar GS, Kim EM, Zavazava N. Demethylation of induced pluripotent stem cells from type 1 diabetic patients enhances differentiation into functional pancreatic β cells. J Biol Chem 2017; 292:14066-14079. [PMID: 28360105 DOI: 10.1074/jbc.m117.784280] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/29/2017] [Indexed: 12/31/2022] Open
Abstract
Type 1 diabetes (T1D) can be managed by transplanting either the whole pancreas or isolated pancreatic islets. However, cadaveric pancreas is scarcely available for clinical use, limiting this approach. As such, there is a great need to identify alternative sources of clinically usable pancreatic tissues. Here, we used induced pluripotent stem (iPS) cells derived from patients with T1D to generate glucose-responsive, insulin-producing cells (IPCs) via 3D culture. Initially, T1D iPS cells were resistant to differentiation, but transient demethylation treatment significantly enhanced IPC yield. The cells responded to high-glucose stimulation by secreting insulin in vitro The shape, size, and number of their granules, as observed by transmission electron microscopy, were identical to those found in cadaveric β cells. When the IPCs were transplanted into immunodeficient mice that had developed streptozotocin-induced diabetes, they promoted a dramatic decrease in hyperglycemia, causing the mice to become normoglycemic within 28 days. None of the mice died or developed teratomas. Because the cells are derived from "self," immunosuppression is not required, providing a much safer and reliable treatment option for T1D patients. Moreover, these cells can be used for drug screening, thereby accelerating drug discovery. In conclusion, our approach eliminates the need for cadaveric pancreatic tissue.
Collapse
Affiliation(s)
- Gohar S Manzar
- From the Department of Internal Medicine and University of Iowa, Iowa City, Iowa; Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa; Veterans Affairs Medical Center, Iowa City, Iowa,; Mayo Clinic College of Medicine, Rochester, Minnesota, and Daejeon 34114, Republic of Korea
| | - Eun-Mi Kim
- From the Department of Internal Medicine and University of Iowa, Iowa City, Iowa; Veterans Affairs Medical Center, Iowa City, Iowa,; Predictive Model Research Center, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Nicholas Zavazava
- From the Department of Internal Medicine and University of Iowa, Iowa City, Iowa; Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa; Veterans Affairs Medical Center, Iowa City, Iowa,.
| |
Collapse
|
86
|
Hohwieler M, Illing A, Hermann PC, Mayer T, Stockmann M, Perkhofer L, Eiseler T, Antony JS, Müller M, Renz S, Kuo CC, Lin Q, Sendler M, Breunig M, Kleiderman SM, Lechel A, Zenker M, Leichsenring M, Rosendahl J, Zenke M, Sainz B, Mayerle J, Costa IG, Seufferlein T, Kormann M, Wagner M, Liebau S, Kleger A. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut 2017; 66:473-486. [PMID: 27633923 PMCID: PMC5534761 DOI: 10.1136/gutjnl-2016-312423] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The generation of acinar and ductal cells from human pluripotent stem cells (PSCs) is a poorly studied process, although various diseases arise from this compartment. DESIGN We designed a straightforward approach to direct human PSCs towards pancreatic organoids resembling acinar and ductal progeny. RESULTS Extensive phenotyping of the organoids not only shows the appropriate marker profile but also ultrastructural, global gene expression and functional hallmarks of the human pancreas in the dish. Upon orthotopic transplantation into immunodeficient mice, these organoids form normal pancreatic ducts and acinar tissue resembling fetal human pancreas without evidence of tumour formation or transformation. Finally, we implemented this unique phenotyping tool as a model to study the pancreatic facets of cystic fibrosis (CF). For the first time, we provide evidence that in vitro, but also in our xenograft transplantation assay, pancreatic commitment occurs generally unhindered in CF. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) activation in mutated pancreatic organoids not only mirrors the CF phenotype in functional assays but also at a global expression level. We also conducted a scalable proof-of-concept screen in CF pancreatic organoids using a set of CFTR correctors and activators, and established an mRNA-mediated gene therapy approach in CF organoids. CONCLUSIONS Taken together, our platform provides novel opportunities to model pancreatic disease and development, screen for disease-rescuing agents and to test therapeutic procedures.
Collapse
Affiliation(s)
- Meike Hohwieler
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Anett Illing
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Patrick C Hermann
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Tobias Mayer
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Marianne Stockmann
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Justin S Antony
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Martin Müller
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Susanne Renz
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Chao-Chung Kuo
- Medical Faculty, IZKF Computational Biology Research Group, RWTH Aachen University, Aachen, Germany
| | - Qiong Lin
- Medical Faculty, Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Markus Breunig
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | | | - André Lechel
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Magdeburg, Germany
| | - Michael Leichsenring
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Division of Medicine, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Martin Zenke
- Medical Faculty, Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Bruno Sainz
- Department of Biochemistry, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, Madrid, Spain
| | - Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Ivan G Costa
- Medical Faculty, IZKF Computational Biology Research Group, RWTH Aachen University, Aachen, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Michael Kormann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Martin Wagner
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| | - Stefan Liebau
- Institute of Neuroanatomy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
87
|
Fu J, Wiraja C, Chong R, Xu C, Wang DA. Real-time and non-invasive monitoring of embryonic stem cell survival during the development of embryoid bodies with smart nanosensor. Acta Biomater 2017; 49:358-367. [PMID: 27845273 DOI: 10.1016/j.actbio.2016.11.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/02/2016] [Accepted: 11/10/2016] [Indexed: 12/27/2022]
Abstract
Embryonic stem cells (ESCs)-derived embryoid body (EB) is a powerful model for the study of early embryonic development and the discovery of therapeutics for tissue regeneration. This article reports a smart nanosensor platform for labeling and tracking the survival and distribution of ESCs during the EB development in a real-time and non-invasive way. Compared with the cell tracker (i.e. DiO) and the green fluorescent protein (GFP), nanosensors provide the homogenous and highly-efficient ESC labeling. Following the internalization, intracellular nanosensors gradually release the non-fluorescent molecules that become fluorescent only in viable cells. This allows a continuous monitoring of ESC survival and distribution during the process of EB formation. Finally, we confirm that nanosensor labeling does not cause the significant influences to biological properties of the ESCs and EBs. STATEMENT OF SIGNIFICANCE The distribution pattern of viable embryonic stem cells (ESCs) within embryoid body (EB) is closely related with the maturation of EBs. Noninvasive and real-time monitoring of viable ESC distribution in EBs would allow researchers to optimize the culturing condition in time during the EB development and to select the suitable EBs for subsequent applications.
Collapse
|
88
|
Akinci B, Sankella S, Gilpin C, Ozono K, Garg A, Agarwal AK. Progeroid syndrome patients with ZMPSTE24 deficiency could benefit when treated with rapamycin and dimethylsulfoxide. Cold Spring Harb Mol Case Stud 2016; 3:a001339. [PMID: 28050601 PMCID: PMC5171694 DOI: 10.1101/mcs.a001339] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Patients with progeroid syndromes such as mandibuloacral dysplasia, type B (MADB) and restrictive dermopathy (RD) harbor mutations in zinc metalloproteinase (ZMPSTE24), an enzyme essential for posttranslational proteolysis of prelamin A to form mature lamin A. Dermal fibroblasts from these patients show increased nuclear dysmorphology and reduced proliferation; however, the efficacy of various pharmacological agents in reversing these cellular phenotypes remains unknown. In this study, fibroblasts from MADB patients exhibited marked nuclear abnormalities and reduced proliferation that improved upon treatment with rapamycin and dimethylsulfoxide but not with other agents, including farnesyl transferase inhibitors. Surprisingly, fibroblasts from an RD patient with a homozygous null mutation in ZMPSTE24, resulting in exclusive accumulation of prelamin A with no lamin A on immunoblotting of cellular lysate, exhibited few nuclear abnormalities and near-normal cellular proliferation. An unbiased proteomic analysis of the cellular lysate from RD fibroblasts revealed a lack of processing of vimentin, a cytoskeletal protein. Interestingly, the assembly of the vimentin microfibrils in MADB fibroblasts improved with rapamycin and dimethylsulfoxide. We conclude that rapamycin and dimethylsulfoxide are beneficial for improving nuclear morphology and cell proliferation of MADB fibroblasts. Data from a single RD patient's fibroblasts also suggest that prelamin A accumulation by itself might not be detrimental and requires additional alterations at the cellular level to manifest the phenotype.
Collapse
Affiliation(s)
- Baris Akinci
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shireesha Sankella
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Christopher Gilpin
- Molecular and Cellular Imaging, Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Anil K Agarwal
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
89
|
Vanhove J, Pistoni M, Welters M, Eggermont K, Vanslembrouck V, Helsen N, Boon R, Najimi M, Sokal E, Collas P, Voncken JW, Verfaillie CM. H3K27me3 Does Not Orchestrate the Expression of Lineage-Specific Markers in hESC-Derived Hepatocytes In Vitro. Stem Cell Reports 2016; 7:192-206. [PMID: 27477635 PMCID: PMC4982990 DOI: 10.1016/j.stemcr.2016.06.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 01/21/2023] Open
Abstract
Although pluripotent stem cells can be differentiated into the hepatocyte lineages, such cells retain an immature phenotype. As the chromatin state of regulatory regions controls spatiotemporal gene expression during development, we evaluated changes in epigenetic histone marks in lineage-specific genes throughout in vitro hepatocyte differentiation from human embryonic stem cells (hESCs). Active acetylation and methylation marks at promoters and enhancers correlated with progressive changes in gene expression. However, repression-associated H3K27me3 marks at these control regions showed an inverse correlation with gene repression during transition from hepatic endoderm to a hepatocyte-like state. Inhibitor of Enhancer of Zeste Homolog 2 (EZH2) reduced H3K27me3 decoration but did not improve hepatocyte maturation. Thus, H3K27me3 at regulatory regions does not regulate transcription and appears dispensable for hepatocyte lineage differentiation of hESCs in vitro. Epigenetic studies to understand hepatocyte differentiation from human PSC Dynamics in histone profile correlate with alterations in gene transcription hESC-derived HLCs have higher H3K27me3 mark at regulatory regions compared with PHHs Reducing H3K27me3 by EZH2 inhibition did not improve hepatocyte differentiation
Collapse
Affiliation(s)
- Jolien Vanhove
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium.
| | - Mariaelena Pistoni
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium.
| | - Marc Welters
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| | - Kristel Eggermont
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| | - Veerle Vanslembrouck
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| | - Nicky Helsen
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| | - Ruben Boon
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, Cliniques St-Luc - Institut de Recherche Expérimentale et Clinique (IREC), Brussels 1200, Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, Cliniques St-Luc - Institut de Recherche Expérimentale et Clinique (IREC), Brussels 1200, Belgium
| | - Philippe Collas
- Faculty of Medicine, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| | - J Willem Voncken
- Department of Molecular Genetics, Maastricht University Medical Centre, 6229 ER Maastricht, the Netherlands
| | - Catherine M Verfaillie
- Department Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
90
|
Swartz EW, Baek J, Pribadi M, Wojta KJ, Almeida S, Karydas A, Gao FB, Miller BL, Coppola G. A Novel Protocol for Directed Differentiation of C9orf72-Associated Human Induced Pluripotent Stem Cells Into Contractile Skeletal Myotubes. Stem Cells Transl Med 2016; 5:1461-1472. [PMID: 27369896 PMCID: PMC5070503 DOI: 10.5966/sctm.2015-0340] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/10/2016] [Indexed: 12/12/2022] Open
Abstract
In the present study, functional skeletal myotubes were efficiently generated from human induced pluripotent stem cells using a small molecule-based approach. Myotubes derived from patients carrying the C9orf72 repeat expansion show no change in differentiation efficiency and normal TDP-43 localization after as many as 120 days in vitro when compared to unaffected controls. The protocol described in this study for the generation of skeletal myotubes from human induced pluripotent stem cells may serve as a valuable tool in drug discovery and modeling of musculoskeletal and neuromuscular diseases. Induced pluripotent stem cells (iPSCs) offer an unlimited resource of cells to be used for the study of underlying molecular biology of disease, therapeutic drug screening, and transplant-based regenerative medicine. However, methods for the directed differentiation of skeletal muscle for these purposes remain scarce and incomplete. Here, we present a novel, small molecule-based protocol for the generation of multinucleated skeletal myotubes using eight independent iPSC lines. Through combinatorial inhibition of phosphoinositide 3-kinase (PI3K) and glycogen synthase kinase 3β (GSK3β) with addition of bone morphogenic protein 4 (BMP4) and fibroblast growth factor 2 (FGF2), we report up to 64% conversion of iPSCs into the myogenic program by day 36 as indicated by MYOG+ cell populations. These cells began to exhibit spontaneous contractions as early as 34 days in vitro in the presence of a serum-free medium formulation. We used this protocol to obtain iPSC-derived muscle cells from frontotemporal dementia (FTD) patients harboring C9orf72 hexanucleotide repeat expansions (rGGGGCC), sporadic FTD, and unaffected controls. iPSCs derived from rGGGGCC carriers contained RNA foci but did not vary in differentiation efficiency when compared to unaffected controls nor display mislocalized TDP-43 after as many as 120 days in vitro. This study presents a rapid, efficient, and transgene-free method for generating multinucleated skeletal myotubes from iPSCs and a resource for further modeling the role of skeletal muscle in amyotrophic lateral sclerosis and other motor neuron diseases. Significance Protocols to produce skeletal myotubes for disease modeling or therapy are scarce and incomplete. The present study efficiently generates functional skeletal myotubes from human induced pluripotent stem cells using a small molecule-based approach. Using this strategy, terminal myogenic induction of up to 64% in 36 days and spontaneously contractile myotubes within 34 days were achieved. Myotubes derived from patients carrying the C9orf72 repeat expansion show no change in differentiation efficiency and normal TDP-43 localization after as many as 120 days in vitro when compared to unaffected controls. This study provides an efficient, novel protocol for the generation of skeletal myotubes from human induced pluripotent stem cells that may serve as a valuable tool in drug discovery and modeling of musculoskeletal and neuromuscular diseases.
Collapse
Affiliation(s)
- Elliot W Swartz
- Interdepartmental Program in Neuroscience, University of California, Los Angeles, Los Angeles, California, USA
| | - Jaeyun Baek
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - Mochtar Pribadi
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - Kevin J Wojta
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna Karydas
- Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Bruce L Miller
- Memory and Aging Center, University of California, San Francisco, San Francisco, California, USA
| | - Giovanni Coppola
- Interdepartmental Program in Neuroscience, University of California, Los Angeles, Los Angeles, California, USA
- Departments of Psychiatry and Neurology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
91
|
Linking the Cell Cycle to Cell Fate Decisions. Trends Cell Biol 2016; 25:592-600. [PMID: 26410405 DOI: 10.1016/j.tcb.2015.07.007] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/20/2015] [Accepted: 07/17/2015] [Indexed: 12/25/2022]
Abstract
Pluripotent stem cells (PSCs) retain the ability to differentiate into a wide range of cell types while undergoing self-renewal. They also exhibit an unusual mode of cell cycle regulation, reflected by a cell cycle structure where G1 and G2 phases are truncated. When individual PSCs are exposed to specification cues, they activate developmental programs and remodel the cell cycle so that the length of G1 and overall cell division times increase. The response of individual stem cells to pro-differentiation signals is strikingly heterogeneous, resulting in asynchronous differentiation. Recent evidence indicates that this phenomenon is due to cell cycle-dependent mechanisms that restrict the initial activation of developmental genes to the G1 phase. This suggests a broad biological mechanism where multipotent cells are 'primed' to initiate cell fate decisions during their transition through G1. Here, I discuss mechanisms underpinning the commitment towards the differentiated state and its relation to the cell cycle.
Collapse
|
92
|
Jiang FX, Morahan G. Insulin-secreting β cells require a post-genomic concept. World J Diabetes 2016; 7:198-208. [PMID: 27226815 PMCID: PMC4873311 DOI: 10.4239/wjd.v7.i10.198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 03/18/2016] [Indexed: 02/05/2023] Open
Abstract
Pancreatic insulin-secreting β cells are essential in maintaining normal glucose homeostasis accomplished by highly specialized transcription of insulin gene, of which occupies up to 40% their transcriptome. Deficiency of these cells causes diabetes mellitus, a global public health problem. Although tremendous endeavors have been made to generate insulin-secreting cells from human pluripotent stem cells (i.e., primitive cells capable of giving rise to all cell types in the body), a regenerative therapy to diabetes has not yet been established. Furthermore, the nomenclature of β cells has become inconsistent, confusing and controversial due to the lack of standardized positive controls of developmental stage-matched in vivo cells. In order to minimize this negative impact and facilitate critical research in this field, a post-genomic concept of pancreatic β cells might be helpful. In this review article, we will briefly describe how β cells were discovered and islet lineage is developed that may help understand the cause of nomenclatural controversy, suggest a post-genomic definition and finally provide a conclusive remark on future research of this pivotal cell.
Collapse
|
93
|
Singh AM, Trost R, Boward B, Dalton S. Utilizing FUCCI reporters to understand pluripotent stem cell biology. Methods 2016; 101:4-10. [PMID: 26404921 PMCID: PMC4801677 DOI: 10.1016/j.ymeth.2015.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/15/2015] [Accepted: 09/20/2015] [Indexed: 11/27/2022] Open
Abstract
The fluorescence ubiquitination cell cycle indicator (FUCCI) system provides a powerful method to evaluate cell cycle mechanisms associated with stem cell self-renewal and cell fate specification. By integrating the FUCCI system into human pluripotent stem cells (hPSCs) it is possible to isolate homogeneous fractions of viable cells representative of all cell cycle phases. This method avoids problems associated with traditional tools used for cell cycle analysis such as synchronizing drugs, elutriation and temperature sensitive mutants. Importantly, FUCCI reporters allow cell cycle events in dynamic systems, such as differentiation, to be evaluated. Initial reports on the FUCCI system focused on its strengths in reporting spatio-temporal aspects of cell cycle events in living cells and developmental models. In this report, we describe approaches that broaden the application of FUCCI reporters in PSCs through incorporation of FACS. This approach allows molecular analysis of the cell cycle in stem cell systems that were not previously possible.
Collapse
Affiliation(s)
- Amar M Singh
- Department of Biochemistry and Molecular Biology, Paul D. Coverdell Center for Biomedical and Health Sciences, The University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Robert Trost
- Department of Biochemistry and Molecular Biology, Paul D. Coverdell Center for Biomedical and Health Sciences, The University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Benjamin Boward
- Department of Biochemistry and Molecular Biology, Paul D. Coverdell Center for Biomedical and Health Sciences, The University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA
| | - Stephen Dalton
- Department of Biochemistry and Molecular Biology, Paul D. Coverdell Center for Biomedical and Health Sciences, The University of Georgia, 500 D.W. Brooks Drive, Athens, GA 30602, USA.
| |
Collapse
|
94
|
PTF1a Activity in Enriched Posterior Foregut Endoderm, but Not Definitive Endoderm, Leads to Enhanced Pancreatic Differentiation in an In Vitro Mouse ESC-Based Model. Stem Cells Int 2016; 2016:6939438. [PMID: 27066080 PMCID: PMC4811216 DOI: 10.1155/2016/6939438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/07/2015] [Accepted: 11/26/2015] [Indexed: 01/22/2023] Open
Abstract
Transcription factors are tools repetitively used by the embryo to generate a variety of lineages. Hence, their context of activation is an important determinant of their ability to specifically trigger certain cell fates, but not others. The context is also consequential when considering directing differentiation of embryonic stem cells (ESCs). In this study, we sought to assess the context of pancreatic transcription factor 1a (PTF1a) activation in reference to its propancreatic effects in mouse ESCs (mESCs). We hypothesized that an enriched endodermal population would respond to PTF1a and trigger the pancreatic program more effectively than a spontaneously differentiated population. Using an in vitro model of pancreas development that we recently established, we found that inducing PTF1a in highly enriched definitive endoderm did not promote pancreatic differentiation but induction in more differentiated endoderm, specifically posterior foregut endoderm, did form pancreatic progenitors. These progenitors never underwent terminal differentiation to endocrine or acinar phenotype. However, a short 3D culture period, prior to PTF1a induction, led to the generation of monohormonal insulin(+) cells and amylase-expressing cells. Our findings suggest that enriched posterior foregut endoderm is competent to respond to PTF1a's propancreatic activity; but a 3D culture environment is essential for terminal differentiation of pancreatic progenitors.
Collapse
|
95
|
Hara J, Tottori J, Anders M, Dadhwal S, Asuri P, Mobed-Miremadi M. Trehalose effectiveness as a cryoprotectant in 2D and 3D cell cultures of human embryonic kidney cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:609-616. [DOI: 10.3109/21691401.2016.1167698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jared Hara
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Jordan Tottori
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Megan Anders
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Smritee Dadhwal
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Prashanth Asuri
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | | |
Collapse
|
96
|
Enrichment of G2/M cell cycle phase in human pluripotent stem cells enhances HDR-mediated gene repair with customizable endonucleases. Sci Rep 2016; 6:21264. [PMID: 26887909 PMCID: PMC4757933 DOI: 10.1038/srep21264] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/15/2016] [Indexed: 12/23/2022] Open
Abstract
Efficient gene editing is essential to fully utilize human pluripotent stem cells (hPSCs) in regenerative medicine. Custom endonuclease-based gene targeting involves two mechanisms of DNA repair: homology directed repair (HDR) and non-homologous end joining (NHEJ). HDR is the preferred mechanism for common applications such knock-in, knock-out or precise mutagenesis, but remains inefficient in hPSCs. Here, we demonstrate that synchronizing synchronizing hPSCs in G2/M with ABT phase increases on-target gene editing, defined as correct targeting cassette integration, 3 to 6 fold. We observed improved efficiency using ZFNs, TALENs, two CRISPR/Cas9, and CRISPR/Cas9 nickase to target five genes in three hPSC lines: three human embryonic stem cell lines, neural progenitors and diabetic iPSCs. neural progenitors and diabetic iPSCs. Reversible synchronization has no effect on pluripotency or differentiation. The increase in on-target gene editing is locus-independent and specific to the cell cycle phase as G2/M phase enriched cells show a 6-fold increase in targeting efficiency compared to cells in G1 phase. Concurrently inhibiting NHEJ with SCR7 does not increase HDR or improve gene targeting efficiency further, indicating that HR is the major DNA repair mechanism after G2/M phase arrest. The approach outlined here makes gene editing in hPSCs a more viable tool for disease modeling, regenerative medicine and cell-based therapies.
Collapse
|
97
|
Gudde AEEG, González-Barriga A, van den Broek WJAA, Wieringa B, Wansink DG. A low absolute number of expanded transcripts is involved in myotonic dystrophy type 1 manifestation in muscle. Hum Mol Genet 2016; 25:1648-62. [PMID: 26908607 PMCID: PMC4805313 DOI: 10.1093/hmg/ddw042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022] Open
Abstract
Muscular manifestation of myotonic dystrophy type 1 (DM1), a common inheritable degenerative multisystem disorder, is mainly caused by expression of RNA from a (CTG·CAG)n-expanded DM1 locus. Here, we report on comparative profiling of expression of normal and expanded endogenous or transgenic transcripts in skeletal muscle cells and biopsies from DM1 mouse models and patients in order to help us in understanding the role of this RNA-mediated toxicity. In tissue of HSALR mice, the most intensely used ‘muscle-only’ model in the DM1 field, RNA from the α-actin (CTG)250 transgene was at least 1000-fold more abundant than that from the Dmpk gene, or the DMPK gene in humans. Conversely, the DMPK transgene in another line, DM500/DMSXL mice, was expressed ∼10-fold lower than the endogenous gene. Temporal regulation of expanded RNA expression differed between models. Onset of expression occurred remarkably late in HSALR myoblasts during in vitro myogenesis whereas Dmpk or DMPK (trans)genes were expressed throughout proliferation and differentiation phases. Importantly, quantification of absolute transcript numbers revealed that normal and expanded Dmpk/DMPK transcripts in mouse models and DM1 patients are low-abundance RNA species. Northern blotting, reverse transcriptase–quantitative polymerase chain reaction, RNA-sequencing and fluorescent in situ hybridization analyses showed that they occur at an absolute number between one and a few dozen molecules per cell. Our findings refine the current RNA dominance theory for DM1 pathophysiology, as anomalous factor binding to expanded transcripts and formation of soluble or insoluble ribonucleoprotein aggregates must be nucleated by only few expanded DMPK transcripts and therefore be a small numbers game.
Collapse
Affiliation(s)
- Anke E E G Gudde
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Anchel González-Barriga
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Walther J A A van den Broek
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
98
|
Julian LM, Carpenedo RL, Rothberg JLM, Stanford WL. Formula G1: Cell cycle in the driver's seat of stem cell fate determination. Bioessays 2016; 38:325-32. [DOI: 10.1002/bies.201500187] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lisa M. Julian
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
| | - Richard L. Carpenedo
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
| | - Janet L. Manias Rothberg
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa ON Canada
| | - William L. Stanford
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa ON Canada
- Department of Biochemistry; Microbiology and Immunology; University of Ottawa; Ottawa ON Canada
- Ottawa Institute of Systems Biology; Ottawa; Ontario Canada
| |
Collapse
|
99
|
Tagliaferri D, De Angelis MT, Russo NA, Marotta M, Ceccarelli M, Del Vecchio L, De Felice M, Falco G. Retinoic Acid Specifically Enhances Embryonic Stem Cell Metastate Marked by Zscan4. PLoS One 2016; 11:e0147683. [PMID: 26840068 PMCID: PMC4740454 DOI: 10.1371/journal.pone.0147683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/07/2016] [Indexed: 11/18/2022] Open
Abstract
Pluripotency confers Embryonic Stem Cells (ESCs) the ability to differentiate in ectoderm, endoderm, and mesoderm derivatives, producing the majority of cell types. Although the majority of ESCs divide without losing pluripotency, it has become evident that ESCs culture consists of multiple cell populations with different degrees of potency that are spontaneously induced in regular ESC culture conditions. Zscan4, a key pluripotency factor, marks ESC subpopulation that is referred to as high-level of pluripotency metastate. Here, we report that in ESC cultures treated with retinoic acid (RA), Zscan4 ESCs metastate is strongly enhanced. In particular, we found that induction of Zscan4 metastate is mediated via RA receptors (RAR-alpha, RAR-beta, and RAR-gamma), and it is dependent on phosphoinositide-3-kinase (PI3K) signaling. Remarkably, Zscan4 metastate induced by RA lacks canonical pluripotency genes Oct3/4 and Nanog but retained both self-renewal and pluripotency capabilities. Finally we demonstrated that the conditional ablation of Zscan4 subpopulation is dispensable for both endoderm and mesoderm but is required for ectoderm lineage. In conclusion, our research provides new insights about the role of RA signaling during ESCs high pluripotency metastate fluctuation.
Collapse
Affiliation(s)
- Daniela Tagliaferri
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore Biogem scarl, Ariano Irpino, Italy
| | | | - Nicola Antonino Russo
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore Biogem scarl, Ariano Irpino, Italy
| | - Maria Marotta
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore Biogem scarl, Ariano Irpino, Italy
| | | | - Luigi Del Vecchio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Mario De Felice
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore Biogem scarl, Ariano Irpino, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Geppino Falco
- Biogem, Istituto di Ricerche Genetiche Gaetano Salvatore Biogem scarl, Ariano Irpino, Italy
- Department of Science, Università degli Studi del Sannio, Benevento, Italy
- Department of Biology, University of Naples “Federico II”, Naples, Italy
- * E-mail:
| |
Collapse
|
100
|
Bartlett ST, Markmann JF, Johnson P, Korsgren O, Hering BJ, Scharp D, Kay TWH, Bromberg J, Odorico JS, Weir GC, Bridges N, Kandaswamy R, Stock P, Friend P, Gotoh M, Cooper DKC, Park CG, O'Connell P, Stabler C, Matsumoto S, Ludwig B, Choudhary P, Kovatchev B, Rickels MR, Sykes M, Wood K, Kraemer K, Hwa A, Stanley E, Ricordi C, Zimmerman M, Greenstein J, Montanya E, Otonkoski T. Report from IPITA-TTS Opinion Leaders Meeting on the Future of β-Cell Replacement. Transplantation 2016; 100 Suppl 2:S1-44. [PMID: 26840096 PMCID: PMC4741413 DOI: 10.1097/tp.0000000000001055] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Stephen T. Bartlett
- Department of Surgery, University of Maryland School of Medicine, Baltimore MD
| | - James F. Markmann
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Paul Johnson
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - David Scharp
- Prodo Laboratories, LLC, Irvine, CA
- The Scharp-Lacy Research Institute, Irvine, CA
| | - Thomas W. H. Kay
- Department of Medicine, St. Vincent’s Hospital, St. Vincent's Institute of Medical Research and The University of Melbourne Victoria, Australia
| | - Jonathan Bromberg
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Jon S. Odorico
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - Gordon C. Weir
- Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Nancy Bridges
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Raja Kandaswamy
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Peter Stock
- Division of Transplantation, University of San Francisco Medical Center, San Francisco, CA
| | - Peter Friend
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Mitsukazu Gotoh
- Department of Surgery, Fukushima Medical University, Fukushima, Japan
| | - David K. C. Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Department of Microbiology and Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Phillip O'Connell
- The Center for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Cherie Stabler
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Shinichi Matsumoto
- National Center for Global Health and Medicine, Tokyo, Japan
- Otsuka Pharmaceutical Factory inc, Naruto Japan
| | - Barbara Ludwig
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden and DZD-German Centre for Diabetes Research, Dresden, Germany
| | - Pratik Choudhary
- Diabetes Research Group, King's College London, Weston Education Centre, London, United Kingdom
| | - Boris Kovatchev
- University of Virginia, Center for Diabetes Technology, Charlottesville, VA
| | - Michael R. Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Coulmbia University Medical Center, New York, NY
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Kristy Kraemer
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Albert Hwa
- Juvenile Diabetes Research Foundation, New York, NY
| | - Edward Stanley
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Monash University, Melbourne, VIC, Australia
| | - Camillo Ricordi
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Mark Zimmerman
- BetaLogics, a business unit in Janssen Research and Development LLC, Raritan, NJ
| | - Julia Greenstein
- Discovery Research, Juvenile Diabetes Research Foundation New York, NY
| | - Eduard Montanya
- Bellvitge Biomedical Research Institute (IDIBELL), Hospital Universitari Bellvitge, CIBER of Diabetes and Metabolic Diseases (CIBERDEM), University of Barcelona, Barcelona, Spain
| | - Timo Otonkoski
- Children's Hospital and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|