1
|
Lee J, Var SR, Chen D, Natera-Rodriguez DE, Hassanipour M, West MD, Low WC, Grande AW, Larocca D. Exosomes derived from highly scalable and regenerative human progenitor cells promote functional improvement in a rat model of ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631793. [PMID: 39829810 PMCID: PMC11741374 DOI: 10.1101/2025.01.07.631793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Globally, there are 15 million stroke patients each year who have significant neurological deficits. Today, there are no treatments that directly address these deficits. With demographics shifting to an older population, the problem is worsening. Therefore, it is crucial to develop feasible therapeutic treatments for stroke. In this study, we tested exosomes derived from embryonic endothelial progenitor cells (eEPC) to assess their therapeutic efficacy in a rat model of ischemic stroke. Importantly, we have developed purification methods aimed at producing robust and scalable exosomes suitable for manufacturing clinical grade therapeutic exosomes. We characterized exosome cargos including RNA-seq, miRNAs targets, and proteomic mass spectrometry analysis, and we found that eEPC-exosomes were enhanced with angiogenic miRNAs (i.e., miR-126), anti-inflammatory miRNA (i.e., miR-146), and anti-apoptotic miRNAs (i.e., miR-21). The angiogenic activity of diverse eEPC-exosomes sourced from a panel of eEPC production lines was assessed in vitro by live-cell vascular tube formation and scratch wound assays, showing that several eEPC-exosomes promoted the proliferation, tube formation, and migration in endothelial cells. We further applied the exosomes systemically in a rat middle cerebral artery occlusion (MCAO) model of stroke and tested for neurological recovery (mNSS) after injury in ischemic animals. The mNSS scores revealed that recovery of sensorimotor functioning in ischemic MCAO rats increased significantly after intravenous administration of eEPC-exosomes and outpaced recovery obtained through treatment with umbilical cord stem cells. Finally, we investigated the potential mechanism of eEPC-exosomes in mitigating ischemic stroke injury and inflammation by the expression of neuronal, endothelial, and inflammatory markers. Taken together, these data support the finding that eEPCs provide a valuable source of exosomes for developing scalable therapeutic products and therapies for stroke and other ischemic diseases.
Collapse
Affiliation(s)
- Jieun Lee
- UniverXome Bioengineering, Inc., (formerly known as AgeX Therapeutics Inc.), Alameda, California, USA
| | - Susanna R. Var
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Derek Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Mohammad Hassanipour
- UniverXome Bioengineering, Inc., (formerly known as AgeX Therapeutics Inc.), Alameda, California, USA
| | - Michael D. West
- UniverXome Bioengineering, Inc., (formerly known as AgeX Therapeutics Inc.), Alameda, California, USA
- LifeCraft Sciences, Inc., Alameda, California, USA
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dana Larocca
- UniverXome Bioengineering, Inc., (formerly known as AgeX Therapeutics Inc.), Alameda, California, USA
- Further Biotechnologies, LLC, Alameda, California, USA
| |
Collapse
|
2
|
Larson A, Natera-Rodriguez DE, Crane A, Larocca D, Low WC, Grande AW, Lee J. Emerging Roles of Exosomes in Stroke Therapy. Int J Mol Sci 2024; 25:6507. [PMID: 38928214 PMCID: PMC11203879 DOI: 10.3390/ijms25126507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke is the number one cause of morbidity in the United States and number two cause of death worldwide. There is a critical unmet medical need for more effective treatments of ischemic stroke, and this need is increasing with the shift in demographics to an older population. Recently, several studies have reported the therapeutic potential of stem cell-derived exosomes as new candidates for cell-free treatment in stoke. This review focuses on the use of stem cell-derived exosomes as a potential treatment tool for stroke patients. Therapy using exosomes can have a clear clinical advantage over stem cell transplantation in terms of safety, cost, and convenience, as well as reducing bench-to-bed latency due to fewer regulatory milestones. In this review article, we focus on (1) the therapeutic potential of exosomes in stroke treatment, (2) the optimization process of upstream and downstream production, and (3) preclinical application in a stroke animal model. Finally, we discuss the limitations and challenges faced by exosome therapy in future clinical applications.
Collapse
Affiliation(s)
- Anthony Larson
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (D.E.N.-R.); (A.C.); (W.C.L.); (A.W.G.)
| | - Dilmareth E. Natera-Rodriguez
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (D.E.N.-R.); (A.C.); (W.C.L.); (A.W.G.)
| | - Andrew Crane
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (D.E.N.-R.); (A.C.); (W.C.L.); (A.W.G.)
| | - Dana Larocca
- DC Biotechnology Consulting, Alameda, CA 94501, USA;
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (D.E.N.-R.); (A.C.); (W.C.L.); (A.W.G.)
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (D.E.N.-R.); (A.C.); (W.C.L.); (A.W.G.)
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jieun Lee
- UniverXome Bioengineering, Inc. (Formerly Known as AgeX Therapeutics Inc.), Alameda, CA 94501, USA
| |
Collapse
|
3
|
Lee J, Sternberg H, Bignone PA, Murai J, Malik NN, West MD, Larocca D. Clonal and Scalable Endothelial Progenitor Cell Lines from Human Pluripotent Stem Cells. Biomedicines 2023; 11:2777. [PMID: 37893151 PMCID: PMC10604251 DOI: 10.3390/biomedicines11102777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) can be used as a renewable source of endothelial cells for treating cardiovascular disease and other ischemic conditions. Here, we present the derivation and characterization of a panel of distinct clonal embryonic endothelial progenitor cells (eEPCs) lines that were differentiated from human embryonic stem cells (hESCs). The hESC line, ESI-017, was first partially differentiated to produce candidate cultures from which eEPCs were cloned. Endothelial cell identity was assessed by transcriptomic analysis, cell surface marker expression, immunocytochemical marker analysis, and functional analysis of cells and exosomes using vascular network forming assays. The transcriptome of the eEPC lines was compared to various adult endothelial lines as well as various non-endothelial cells including both adult and embryonic origins. This resulted in a variety of distinct cell lines with functional properties of endothelial cells and strong transcriptomic similarity to adult endothelial primary cell lines. The eEPC lines, however, were distinguished from adult endothelium by their novel pattern of embryonic gene expression. We demonstrated eEPC line scalability of up to 80 population doublings (pd) and stable long-term expansion of over 50 pd with stable angiogenic properties at late passage. Taken together, these data support the finding that hESC-derived clonal eEPC lines are a potential source of scalable therapeutic cells and cell products for treating cardiovascular disease. These eEPC lines offer a highly promising resource for the development of further preclinical studies aimed at therapeutic interventions.
Collapse
Affiliation(s)
- Jieun Lee
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - Paola A. Bignone
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - James Murai
- Advanced Cell Technology, Alameda, CA 94502, USA
| | - Nafees N. Malik
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | | | - Dana Larocca
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| |
Collapse
|
4
|
West MD, Sternberg H, Labat I, Janus J, Chapman KB, Malik NN, de Grey ADNJ, Larocca D. Toward a unified theory of aging and regeneration. Regen Med 2019; 14:867-886. [DOI: 10.2217/rme-2019-0062] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the antagonistic pleiotropy theory of mammalian aging. Accordingly, changes in gene expression following the pluripotency transition, and subsequent transitions such as the embryonic–fetal transition, while providing tumor suppressive and antiviral survival benefits also result in a loss of regenerative potential leading to age-related fibrosis and degenerative diseases. However, reprogramming somatic cells to pluripotency demonstrates the possibility of restoring telomerase and embryonic regeneration pathways and thus reversing the age-related decline in regenerative capacity. A unified model of aging and loss of regenerative potential is emerging that may ultimately be translated into new therapeutic approaches for establishing induced tissue regeneration and modulation of the embryo-onco phenotype of cancer.
Collapse
Affiliation(s)
| | | | - Ivan Labat
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
| | | | | | - Nafees N Malik
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
- Juvenescence Ltd, London, UK
| | - Aubrey DNJ de Grey
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
- SENS Research Foundation, Mountain View, CA 94041, USA
| | | |
Collapse
|
5
|
West MD, Chang CF, Larocca D, Li J, Jiang J, Sim P, Labat I, Chapman KB, Wong KE, Nicoll J, Van Kanegan MJ, de Grey ADNJ, Nasonkin IO, Stahl A, Sternberg H. Clonal derivation of white and brown adipocyte progenitor cell lines from human pluripotent stem cells. Stem Cell Res Ther 2019; 10:7. [PMID: 30616682 PMCID: PMC6323697 DOI: 10.1186/s13287-018-1087-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The role of brown fat in non-shivering thermogenesis and the discovery of brown fat depots in adult humans has made it the subject of intense research interest. A renewable source of brown adipocyte (BA) progenitors would be highly valuable for research and therapy. Directed differentiation of human pluripotent stem (hPS) cells to white or brown adipocytes is limited by lack of cell purity and scalability. Here we describe an alternative approach involving the identification of clonal self-renewing human embryonic progenitor (hEP) cell lines following partial hPS cell differentiation and selection of scalable clones. METHODS We screened a diverse panel of hPS cell-derived clonal hEP cell lines for adipocyte markers following growth in adipocyte differentiation medium. The transcriptome of the human hES-derived clonal embryonic progenitor cell lines E3, C4ELS5.1, NP88, and NP110 representing three class of definitive adipocyte progenitors were compared to the relatively non-adipogenic line E85 and adult-derived BAT and SAT-derived cells using gene expression microarrays, RT-qPCR, metabolic analysis and immunocytochemistry. Differentiation conditions were optimized for maximal UCP1 expression. RESULTS Many of the differentiated hEP cell lines expressed the adipocyte marker, FAPB4, but only a small subset expressed definitive adipocyte markers including brown adipocyte marker, UCP1. Class I cells (i.e., E3) expressed CITED1, ADIPOQ, and C19orf80 but little to no UCP1. Class II (i.e., C4ELS5.1) expressed CITED1 and UCP1 but little ADIPOQ and LIPASIN. Class III (i.e., NP88, NP110) expressed CITED1, ADIPOQ, C19orf80, and UCP1 in a similar manner as fetal BAT-derived (fBAT) cells. Differentiated NP88 and NP110 lines were closest to fBAT cells morphologically in adiponectin and uncoupling protein expression. But they were more metabolically active than fBAT cells, had higher levels of 3-hydroxybutyrate, and lacked expression of fetal/adult marker, COX7A1. The hEP BA progenitor lines were scalable to 17 passages without loss of differentiation capacity and could be readily rederived. CONCLUSIONS Taken together, these data demonstrate that self-renewing adipocyte progenitor cells can be derived from hES cells and that they are functionally like BAT cells but with unique properties that might be advantageous for basic research and for development of cell-based treatments for metabolic diseases.
Collapse
Affiliation(s)
- Michael D. West
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ching-Fang Chang
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Dana Larocca
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jie Li
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jianjie Jiang
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Pamela Sim
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ivan Labat
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Karen B. Chapman
- 0000 0001 2171 9311grid.21107.35Johns Hopkins University, Baltimore, MD 21218 USA
| | - Kari E. Wong
- grid.429438.0Metabolon Inc., Morrisville, NC 27560 USA
| | - James Nicoll
- grid.422945.cZen-Bio, Inc., Research Triangle Park, NC 27709 USA
| | | | - Aubrey D. N. J. de Grey
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA ,SENS Research Foundation, Mountain View, CA 94041 USA
| | | | - Andreas Stahl
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| |
Collapse
|
6
|
Amzaleg Y, Ji J, Kittivanichkul D, E Törnqvist A, Windahl S, Sabag E, Khalid AB, Sternberg H, West M, Katzenellenbogen JA, Krum SA, Chimge NO, Schones DE, Gabet Y, Ohlsson C, Frenkel B. Estrogens and selective estrogen receptor modulators differentially antagonize Runx2 in ST2 mesenchymal progenitor cells. J Steroid Biochem Mol Biol 2018; 183:10-17. [PMID: 29751107 PMCID: PMC6128776 DOI: 10.1016/j.jsbmb.2018.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/05/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022]
Abstract
Estrogens attenuate bone turnover by inhibiting both osteoclasts and osteoblasts, in part through antagonizing Runx2. Apparently conflicting, stimulatory effects in osteoblast lineage cells, however, sway the balance between bone resorption and bone formation in favor of the latter. Consistent with this dualism, 17ß-estradiol (E2) both stimulates and inhibits Runx2 in a locus-specific manner, and here we provide evidence for such locus-specific regulation of Runx2 by E2 in vivo. We also demonstrate dual, negative and positive, regulation of Runx2-driven alkaline phosphatase (ALP) activity by increasing E2 concentrations in ST2 osteoblast progenitor cells. We further compared the effects of E2 to those of the Selective Estrogen Receptor Modulators (SERMs) raloxifene (ral) and lasofoxifene (las) and the phytoestrogen puerarin. We found that E2 at the physiological concentrations of 0.1-1 nM, as well as ral and las, but not puerarin, antagonize Runx2-driven ALP activity. At ≥10 nM, E2 and puerarin, but not ral or las, stimulate ALP relative to the activity measured at 0.1-1 nM. Contrasting the difference between E2 and SERMs in ST2 cells, they all shared a similar dose-response profile when inhibiting pre-osteoclast proliferation. That ral and las poorly mimic the locus- and concentration-dependent effects of E2 in mesenchymal progenitor cells may help explain their limited clinical efficacy.
Collapse
Affiliation(s)
- Yonatan Amzaleg
- Center of Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA; Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Jie Ji
- Departments of Biochemistry and Molecular Medicine, Los Angeles, CA, USA; Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | | | - Anna E Törnqvist
- Center for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sara Windahl
- Center for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elias Sabag
- Center of Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA; Departments of Biochemistry and Molecular Medicine, Los Angeles, CA, USA
| | - Aysha B Khalid
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hal Sternberg
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, CA, USA
| | - Michael West
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, CA, USA
| | | | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Dustin E Schones
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Baruch Frenkel
- Center of Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA; Departments of Biochemistry and Molecular Medicine, Los Angeles, CA, USA; Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; Departments of Orthopedic Surgery, Los Angeles, CA, USA.
| |
Collapse
|
7
|
West MD, Labat I, Sternberg H, Larocca D, Nasonkin I, Chapman KB, Singh R, Makarev E, Aliper A, Kazennov A, Alekseenko A, Shuvalov N, Cheskidova E, Alekseev A, Artemov A, Putin E, Mamoshina P, Pryanichnikov N, Larocca J, Copeland K, Izumchenko E, Korzinkin M, Zhavoronkov A. Use of deep neural network ensembles to identify embryonic-fetal transition markers: repression of COX7A1 in embryonic and cancer cells. Oncotarget 2017; 9:7796-7811. [PMID: 29487692 PMCID: PMC5814259 DOI: 10.18632/oncotarget.23748] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022] Open
Abstract
Here we present the application of deep neural network (DNN) ensembles trained on transcriptomic data to identify the novel markers associated with the mammalian embryonic-fetal transition (EFT). Molecular markers of this process could provide important insights into regulatory mechanisms of normal development, epimorphic tissue regeneration and cancer. Subsequent analysis of the most significant genes behind the DNNs classifier on an independent dataset of adult-derived and human embryonic stem cell (hESC)-derived progenitor cell lines led to the identification of COX7A1 gene as a potential EFT marker. COX7A1, encoding a cytochrome C oxidase subunit, was up-regulated in post-EFT murine and human cells including adult stem cells, but was not expressed in pre-EFT pluripotent embryonic stem cells or their in vitro-derived progeny. COX7A1 expression level was observed to be undetectable or low in multiple sarcoma and carcinoma cell lines as compared to normal controls. The knockout of the gene in mice led to a marked glycolytic shift reminiscent of the Warburg effect that occurs in cancer cells. The DNN approach facilitated the elucidation of a potentially new biomarker of cancer and pre-EFT cells, the embryo-onco phenotype, which may potentially be used as a target for controlling the embryonic-fetal transition.
Collapse
Affiliation(s)
| | - Ivan Labat
- AgeX Therapeutics, Inc., Alameda, CA, USA
| | | | | | | | | | | | - Eugene Makarev
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Alex Aliper
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Andrey Kazennov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Andrey Alekseenko
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Innopolis University, Innoplis, Russia
| | - Nikolai Shuvalov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Evgenia Cheskidova
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksandr Alekseev
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Artem Artemov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Evgeny Putin
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Computer Technologies Lab, ITMO University, St. Petersburg, Russia
| | - Polina Mamoshina
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Nikita Pryanichnikov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | | | | | - Evgeny Izumchenko
- Johns Hopkins University, School of Medicine, Department of Otolaryngology-Head and Neck Cancer Research, Baltimore, MD, USA
| | - Mikhail Korzinkin
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,The Biogerontology Research Foundation, Trevissome Park, Truro, UK
| |
Collapse
|
8
|
West MD, Nasonkin I, Larocca D, Chapman KB, Binette F, Sternberg H. Adult Versus Pluripotent Stem Cell-Derived Mesenchymal Stem Cells: The Need for More Precise Nomenclature. CURRENT STEM CELL REPORTS 2016; 2:299-303. [PMID: 27547711 PMCID: PMC4972883 DOI: 10.1007/s40778-016-0060-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The complexity of human pluripotent stem cell (hPSC) fate represents both opportunity and challenge. In theory, all somatic cell types can be differentiated from hPSCs, opening the door to many opportunities in transplant medicine. However, such clinical applications require high standards of purity and identity, that challenge many existing protocols. This underscores the need for increasing precision in the description of cell identity during hPSC differentiation. We highlight one salient example, namely, the numerous published reports of hPSC-derived mesenchymal stem cells (MSCs). We suggest that many of these reports likely represent an improper use of certain cluster of differentiation (CD) antigens in defining bone marrow-derived MSCs. Instead, most such hPSC-derived mesenchymal cells are likely a complex mixture of embryonic anlagen, primarily of diverse mesodermal and neural crest origins, making precise identification, reproducible manufacture, and uniform differentiation difficult to achieve. We describe a potential path forward that may provide more precision in nomenclature, and cells with higher purity and identity for potential therapeutic use.
Collapse
Affiliation(s)
- Michael D West
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501 USA
| | - Igor Nasonkin
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501 USA
| | - David Larocca
- ReCyte Therapeutics, 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Karen B Chapman
- OncoCyte Corporation, 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Francois Binette
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501 USA ; OrthoCyte Corporation, 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Hal Sternberg
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501 USA
| |
Collapse
|
9
|
West MD, Binette F, Larocca D, Chapman KB, Irving C, Sternberg H. The germline/soma dichotomy: implications for aging and degenerative disease. Regen Med 2016; 11:331-4. [DOI: 10.2217/rme-2015-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human somatic cells are mortal due in large part to telomere shortening associated with cell division. Limited proliferative capacity may, in turn, limit response to injury and may play an important role in the etiology of age-related pathology. Pluripotent stem cells cultured in vitro appear to maintain long telomere length through relatively high levels of telomerase activity. We propose that the induced reversal of cell aging by transcriptional reprogramming, or alternatively, human embryonic stem cells engineered to escape immune surveillance, are effective platforms for the industrial-scale manufacture of young cells for the treatment of age-related pathologies. Such cell-based regenerative therapies will require newer manufacturing and delivery technologies to insure highly pure, identified and potent pluripotency-based therapeutic formulations.
Collapse
Affiliation(s)
- Michael D West
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501, USA
| | | | | | | | | | - Hal Sternberg
- BioTime, Inc., 1010 Atlantic Ave., Alameda, CA 94501, USA
| |
Collapse
|
10
|
Yu JL, Adisetiyo H, Little GH, Vangsness CT, Jiang J, Sternberg H, West MD, Frenkel B. Initial Characterization of Osteoblast Differentiation and Loss of RUNX2 Stability in the Newly Established SK11 Human Embryonic Stem Cell-Derived Cell Line. J Cell Physiol 2015; 230:237-41. [PMID: 25160731 DOI: 10.1002/jcp.24773] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 08/22/2014] [Indexed: 11/11/2022]
Abstract
We describe a novel model for investigation of genetically normal human osteoblasts in culture. SK11 is a clonal progenitor cell line derived from human embryonic stem cells. Initially selected based on the expression of chondrogenic markers when differentiated in micromass culture, SK11 cells display typical mRNA expression patterns of bone phenotypic genes under osteogenic conditions. These include osterix, α1(I) collagen, alkaline phosphatase, osteonectin, osteopontin, and osteocalcin. Similar to well-characterized murine osteoblast cultures, the osteoblast master regulator RUNX2 was present during the first few days after plating, but the protein disappeared during the first week of culture. Loss of RUNX2 expression is considered an important regulatory feature for osteoblast maturation. Indeed, following ∼2 weeks of differentiation, SK11 cultures exhibited robust calcium deposition, evidenced by alizarin red staining. We also introduced a lentiviral vector encoding doxycycline (dox)-inducible FLAG-tagged RUNX2 into SK11 cells. Dox-mediated enhancement of RUNX2 expression resulted in accelerated mineralization, which was further increased by co-treatment with BMP-2. Like the endogenous RUNX2, expression of the virally coded FLAG-RUNX2 was lost during the first week of culture despite persistent dox treatment. By following RUNX2 decay after dox withdrawal from day-5 versus day-3 cultures, we demonstrated a developmentally regulated decrease in RUNX2 stability. Availability of culture models for molecular investigation of genetically normal human osteoblasts is important because differences between murine and human osteoblasts, demonstrated here by the regulation of matrix Gla Protein, may have significant biomedical implications.
Collapse
Affiliation(s)
- Jia-Li Yu
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California.,Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Helty Adisetiyo
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California.,Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Gillian H Little
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California.,Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - C Thomas Vangsness
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, California
| | - Jianjie Jiang
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, California
| | - Hal Sternberg
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, California
| | - Michael D West
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, California
| | - Baruch Frenkel
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California.,Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Department of Orthopaedic Surgery, University of Southern California, Los Angeles, California
| |
Collapse
|
11
|
Bignone PA, Krupa RA, West MD, Larocca D. Selection of Phage Display Peptides Targeting Human Pluripotent Stem Cell-Derived Progenitor Cell Lines. Methods Mol Biol 2014; 1357:269-83. [PMID: 25410289 DOI: 10.1007/7651_2014_144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The ability of human pluripotent stem cells (hPS) to both self-renew and differentiate into virtually any cell type makes them a promising source of cells for cell-based regenerative therapies. However, stem cell identity, purity, and scalability remain formidable challenges that need to be overcome for translation of pluripotent stem cell research into clinical applications. Directed differentiation from hPS cells is inefficient and residual contamination with pluripotent cells that have the potential to form tumors remains problematic. The derivation of scalable (self-renewing) embryonic progenitor stem cell lines offers a solution because they are well defined and clonally pure. Clonally pure progenitor stem cell lines also provide a means for identifying cell surface targeting reagents that are useful for identification, tracking, and repeated derivation of the corresponding progenitor stem cell types from additional hPS cell sources. Such stem cell targeting reagents can then be applied to the manufacture of genetically diverse banks of human embryonic progenitor cell lines for drug screening, disease modeling, and cell therapy. Here we present methods to identify human embryonic progenitor stem cell targeting peptides by selection of phage display libraries on clonal embryonic progenitor cell lines and demonstrate their use for targeting quantum dots (Qdots) for stem cell labeling.
Collapse
Affiliation(s)
| | | | - Michael D West
- BioTime Inc., 1301 Harbor Bay Parkway, Alameda, CA, 94502, USA
| | | |
Collapse
|
12
|
Sternberg H, Jiang J, Sim P, Kidd J, Janus J, Rinon A, Edgar R, Shitrit A, Larocca D, Chapman KB, Binette F, West MD. Human embryonic stem cell-derived neural crest cells capable of expressing markers of osteochondral or meningeal-choroid plexus differentiation. Regen Med 2014; 9:53-66. [PMID: 24351006 DOI: 10.2217/rme.13.86] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
AIMS The transcriptome and fate potential of three diverse human embryonic stem cell-derived clonal embryonic progenitor cell lines with markers of cephalic neural crest are compared when differentiated in the presence of combinations of TGFβ3, BMP4, SCF and HyStem-C matrices. MATERIALS & METHODS The cell lines E69 and T42 were compared with MEL2, using gene expression microarrays, immunocytochemistry and ELISA. RESULTS In the undifferentiated progenitor state, each line displayed unique markers of cranial neural crest including TFAP2A and CD24; however, none expressed distal HOX genes including HOXA2 or HOXB2, or the mesenchymal stem cell marker CD74. The lines also showed diverse responses when differentiated in the presence of exogenous BMP4, BMP4 and TGFβ3, SCF, and SCF and TGFβ3. The clones E69 and T42 showed a profound capacity for expression of endochondral ossification markers when differentiated in the presence of BMP4 and TGFβ3, choroid plexus markers in the presence of BMP4 alone, and leptomeningeal markers when differentiated in SCF without TGFβ3. CONCLUSION The clones E69 and T42 may represent a scalable source of primitive cranial neural crest cells useful in the study of cranial embryology, and potentially cell-based therapy.
Collapse
Affiliation(s)
- Hal Sternberg
- BioTime, Inc., 1301 Harbor Bay, Parkway, Alameda, CA 94502, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hussain W, Moens N, Veraitch FS, Hernandez D, Mason C, Lye GJ. Reproducible culture and differentiation of mouse embryonic stem cells using an automated microwell platform. Biochem Eng J 2013; 77:246-257. [PMID: 23956681 PMCID: PMC3741632 DOI: 10.1016/j.bej.2013.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 04/16/2013] [Accepted: 05/16/2013] [Indexed: 02/05/2023]
Abstract
We describe an automated platform for hands-free ESC expansion and differentiation. Key bioprocess variables were investigated to optimize culture inductions. Cell growth was more consistent with automated ESC expansion than manual culture. ESCs expanded on the automated platform maintained high levels of pluripotency. Cells expressed βIII-tubulin after successful automated neuronal differentiation.
The use of embryonic stem cells (ESCs) and their progeny in high throughput drug discovery and regenerative medicine will require production at scale of well characterized cells at an appropriate level of purity. The adoption of automated bioprocessing techniques offers the possibility to overcome the lack of consistency and high failure rates seen with current manual protocols. To build the case for increased use of automation this work addresses the key question: “can an automated system match the quality of a highly skilled and experienced person working manually?” To answer this we first describe an integrated automation platform designed for the ‘hands-free’ culture and differentiation of ESCs in microwell formats. Next we outline a framework for the systematic investigation and optimization of key bioprocess variables for the rapid establishment of validatable Standard Operating Procedures (SOPs). Finally the experimental comparison between manual and automated bioprocessing is exemplified by expansion of the murine Oct-4-GiP ESC line over eight sequential passages with their subsequent directed differentiation into neural precursors. Our results show that ESCs can be effectively maintained and differentiated in a highly reproducible manner by the automated system described. Statistical analysis of the results for cell growth over single and multiple passages shows up to a 3-fold improvement in the consistency of cell growth kinetics with automated passaging. The quality of the cells produced was evaluated using a panel of biological markers including cell growth rate and viability, nutrient and metabolite profiles, changes in gene expression and immunocytochemistry. Automated processing of the ESCs had no measurable negative effect on either their pluripotency or their ability to differentiate into the three embryonic germ layers. Equally important is that over a 6-month period of culture without antibiotics in the medium, we have not had any cases of culture contamination. This study thus confirms the benefits of adopting automated bioprocess routes to produce cells for therapy and for use in basic discovery research.
Collapse
Affiliation(s)
- Waqar Hussain
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | | | | | | | | | | |
Collapse
|
14
|
LifeMap Discovery™: the embryonic development, stem cells, and regenerative medicine research portal. PLoS One 2013; 8:e66629. [PMID: 23874394 PMCID: PMC3714290 DOI: 10.1371/journal.pone.0066629] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 04/29/2013] [Indexed: 11/19/2022] Open
Abstract
LifeMap Discovery™ provides investigators with an integrated database of embryonic development, stem cell biology and regenerative medicine. The hand-curated reconstruction of cell ontology with stem cell biology; including molecular, cellular, anatomical and disease-related information, provides efficient and easy-to-use, searchable research tools. The database collates in vivo and in vitro gene expression and guides translation from in vitro data to the clinical utility, and thus can be utilized as a powerful tool for research and discovery in stem cell biology, developmental biology, disease mechanisms and therapeutic discovery. LifeMap Discovery is freely available to academic nonprofit institutions at http://discovery.lifemapsc.com.
Collapse
|
15
|
Bignone PA, Krupa RA, Sternberg H, Funk WD, Snyder EY, West MD, Larocca D. Identification of human embryonic progenitor cell targeting peptides using phage display. PLoS One 2013; 8:e58200. [PMID: 23483995 PMCID: PMC3587414 DOI: 10.1371/journal.pone.0058200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 02/04/2013] [Indexed: 11/18/2022] Open
Abstract
Human pluripotent stem (hPS) cells are capable of differentiation into derivatives of all three primary embryonic germ layers and can self-renew indefinitely. They therefore offer a potentially scalable source of replacement cells to treat a variety of degenerative diseases. The ability to reprogram adult cells to induced pluripotent stem (iPS) cells has now enabled the possibility of patient-specific hPS cells as a source of cells for disease modeling, drug discovery, and potentially, cell replacement therapies. While reprogramming technology has dramatically increased the availability of normal and diseased hPS cell lines for basic research, a major bottleneck is the critical unmet need for more efficient methods of deriving well-defined cell populations from hPS cells. Phage display is a powerful method for selecting affinity ligands that could be used for identifying and potentially purifying a variety of cell types derived from hPS cells. However, identification of specific progenitor cell-binding peptides using phage display may be hindered by the large cellular heterogeneity present in differentiating hPS cell populations. We therefore tested the hypothesis that peptides selected for their ability to bind a clonal cell line derived from hPS cells would bind early progenitor cell types emerging from differentiating hPS cells. The human embryonic stem (hES) cell-derived embryonic progenitor cell line, W10, was used and cell-targeting peptides were identified. Competition studies demonstrated specificity of peptide binding to the target cell surface. Efficient peptide targeted cell labeling was accomplished using multivalent peptide-quantum dot complexes as detected by fluorescence microscopy and flow cytometry. The cell-binding peptides were selective for differentiated hPS cells, had little or no binding on pluripotent cells, but preferential binding to certain embryonic progenitor cell lines and early endodermal hPS cell derivatives. Taken together these data suggest that selection of phage display libraries against a clonal progenitor stem cell population can be used to identify progenitor stem cell targeting peptides. The peptides may be useful for monitoring hPS cell differentiation and for the development of cell enrichment procedures to improve the efficiency of directed differentiation toward clinically relevant human cell types.
Collapse
Affiliation(s)
- Paola A. Bignone
- Mandala Biosciences LLC, San Diego, California, United States of America
| | - Rachel A. Krupa
- Mandala Biosciences LLC, San Diego, California, United States of America
| | - Hal Sternberg
- BioTime Inc., Alameda, California, United States of America
| | - Walter D. Funk
- BioTime Inc., Alameda, California, United States of America
| | - Evan Y. Snyder
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | | | - David Larocca
- Mandala Biosciences LLC, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Sternberg H, Janus J, West MD. Defining cell-matrix combination products in the era of pluripotency. BIOMATTER 2013; 3:24496. [PMID: 23567186 PMCID: PMC3732326 DOI: 10.4161/biom.24496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human pluripotent stem (hPS) cells provide an attractive opportunity for the manufacture of a wide array of therapeutic cell types. The challenges to commercialization include the thousand-fold diversity of cell types emerging from hPS cells and the associated difficulties in validating processes to reliably generate cells with precise identity and purity. Improved methods of controlling the dosage and migration of hPS-derived cells in solid tissues are also needed. To directly address these issues, we clonally expanded proliferating lineages of cells that were intermediate in regard to their state of differentiation between hPS and terminally differentiated cells. These cells called monoclonal embryonic progenitors (hEP), are expandable mortal lineages with diverse site-specific homeobox gene expression and multipotentiality. In this review, we discuss methods of generating combination products wherein the fate space of precisely identified monoclonal hEP cells is mapped by differentiating the cells in vitro in HyStem-3D bead arrays in the presence of diverse growth factors. This combination of discovery processes has the potential to translate directly into cell-matrix formulations that can be used to generate pre-clinical data leading to human clinical trials and potentially new medical therapies.
Collapse
|
17
|
Sternberg H, Kidd J, Murai JT, Jiang J, Rinon A, Erickson IE, Funk WD, Wang Q, Chapman KB, Vangsness CT, West MD. Seven diverse human embryonic stem cell-derived chondrogenic clonal embryonic progenitor cell lines display site-specific cell fates. Regen Med 2012; 8:125-44. [PMID: 23249126 DOI: 10.2217/rme.12.117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
AIM The transcriptomes of seven diverse clonal human embryonic progenitor cell lines with chondrogenic potential were compared with that of bone marrow-derived mesenchymal stem cells (MSCs). MATERIALS & METHODS The cell lines 4D20.8, 7PEND24, 7SMOO32, E15, MEL2, SK11 and SM30 were compared with MSCs using immunohistochemical methods, gene expression microarrays and quantitative real-time PCR. RESULTS In the undifferentiated progenitor state, each line displayed unique combinations of site-specific markers, including AJAP1, ALDH1A2, BMP5, BARX1, HAND2, HOXB2, LHX1, LHX8, PITX1, TBX15 and ZIC2, but none of the lines expressed the MSC marker CD74. The lines showed diverse responses when differentiated in the presence of combinations of TGF-β3, BMP2, 4, 6 and 7 and GDF5, with the lines 4D20.8, SK11, SM30 and MEL2 showing osteogenic markers in some differentiation conditions. The line 7PEND24 showed evidence of regenerating articular cartilage and, in some conditions, markers of tendon differentiation. CONCLUSION The scalability of site-specific clonal human embryonic stem cell-derived embryonic progenitor cell lines may provide novel models for the study of differentiation and methods for preparing purified and identified cells types for use in therapy.
Collapse
Affiliation(s)
- Hal Sternberg
- BioTime, Inc., 1301 Harbor Bay Parkway, Alameda, CA 94502, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Grosse-Gehling P, Fargeas CA, Dittfeld C, Garbe Y, Alison MR, Corbeil D, Kunz-Schughart LA. CD133 as a biomarker for putative cancer stem cells in solid tumours: limitations, problems and challenges. J Pathol 2012; 229:355-78. [DOI: 10.1002/path.4086] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/30/2012] [Accepted: 08/04/2012] [Indexed: 12/11/2022]
Affiliation(s)
- Philipp Grosse-Gehling
- Tumor Pathophysiology, OncoRay, National Center for Radiation Research in Oncology; Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| | - Christine A Fargeas
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD); Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| | - Claudia Dittfeld
- Tumor Pathophysiology, OncoRay, National Center for Radiation Research in Oncology; Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| | - Yvette Garbe
- Tumor Pathophysiology, OncoRay, National Center for Radiation Research in Oncology; Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| | - Malcolm R Alison
- Blizard Institute; Barts and The London School of Medicine and Dentistry; London; UK
| | - Denis Corbeil
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD); Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| | - Leoni A Kunz-Schughart
- Tumor Pathophysiology, OncoRay, National Center for Radiation Research in Oncology; Dresden University of Technology; Fetscherstrasse 74; 01307; Dresden; Germany
| |
Collapse
|
19
|
Sternberg H, Murai JT, Erickson IE, Funk WD, Das S, Wang Q, Snyder E, Chapman KB, Vangsness CT, West MD. A human embryonic stem cell-derived clonal progenitor cell line with chondrogenic potential and markers of craniofacial mesenchyme. Regen Med 2012; 7:481-501. [PMID: 22519755 DOI: 10.2217/rme.12.29] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIMS We screened 100 diverse human embryonic stem-derived progenitor cell lines to identify novel lines with chondrogenic potential. MATERIALS & METHODS The 4D20.8 cell line was compared with mesenchymal stem cells and dental pulp stem cells by assessing osteochondral markers using immunohistochemical methods, gene expression microarrays, quantitative real-time PCR and in vivo repair of rat articular condyles. RESULTS 4D20.8 expressed the site-specific gene markers LHX8 and BARX1 and robustly upregulated chondrocyte markers upon differentiation. Differentiated 4D20.8 cells expressed relatively low levels of COL10A1 and lacked IHH and CD74 expression. Transplantation of 4D20.8 cells into experimentally induced defects in the femoral condyle of athymic rats resulted in cartilage and bone differentiation approximating that of the original tissue architecture. Relatively high COL2A1 and minimal COL10A1 expression occurred during differentiation in HyStem-C hydrogel with TGF-β3 and GDF-5. CONCLUSION Human embryonic stem cell-derived embryonic progenitor cell lines may provide a novel means of generating purified site-specific osteochondral progenitor cell lines that are useful in research and therapy.
Collapse
|
20
|
Chemical biology in stem cell research. Arch Pharm Res 2012; 35:281-97. [PMID: 22370782 DOI: 10.1007/s12272-012-0208-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 12/07/2011] [Accepted: 12/07/2011] [Indexed: 10/28/2022]
Abstract
Stem cells are offering a considerable range of prospects to the biomedical research including novel platforms for disease models and drug discovery tools to cell transplantation and regenerative therapies. However, there are several obstacles to overcome to bring these potentials into reality. First, robust methods to maintain stem cells in the pluripotent state should be established and factors that are required to direct stem cell fate into a particular lineage should be elucidated. Second, both allogeneic rejection following transplantation and limited cell availability issues must be circumvented. These challenges are being addressed, at least in part, through the identification of a group of chemicals (small molecules) that possess novel activities on stem cell biology. For example, small molecules can be used both in vitro and/or in vivo as tools to promote proliferation of stem cells (self-renewal), to direct stem cells to a lineage specific patterns (differentiation), or to reprogram somatic cells to a more undifferentiated state (de-differentiation or reprogramming). These molecules, in turn, have provided new insights into the signaling mechanisms that regulate stem cell biology, and may eventually lead to effective therapies in regenerative medicine. In this review, we will introduce recent findings with regards to small molecules and their impact on stem cell self-renewal and differentiation.
Collapse
|
21
|
Teichroeb JH, Betts DH, Vaziri H. Suppression of the imprinted gene NNAT and X-chromosome gene activation in isogenic human iPS cells. PLoS One 2011; 6:e23436. [PMID: 22022350 PMCID: PMC3192059 DOI: 10.1371/journal.pone.0023436] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 07/18/2011] [Indexed: 01/21/2023] Open
Abstract
Genetic comparison between human embryonic stem cells and induced pluripotent stem cells has been hampered by genetic variation. To solve this problem, we have developed an isogenic system that allows direct comparison of induced pluripotent stem cells (hiPSCs) to their genetically matched human embryonic stem cells (hESCs). We show that hiPSCs have a highly similar transcriptome to hESCs. Global transcriptional profiling identified 102–154 genes (>2 fold) that showed a difference between isogenic hiPSCs and hESCs. A stringent analysis identified NNAT as a key imprinted gene that was dysregulated in hiPSCs. Furthermore, a disproportionate number of X-chromosome localized genes were over-expressed in female hiPSCs. Our results indicate that despite a remarkably close transcriptome to hESCs, isogenic hiPSCs have alterations in imprinting and regulation of X-chromosome genes.
Collapse
Affiliation(s)
- Jonathan H. Teichroeb
- Ontario Cancer Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Dean H. Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Homayoun Vaziri
- Ontario Cancer Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemical Control of Stem Cell Fate and Developmental Potential. Angew Chem Int Ed Engl 2010; 50:200-42. [DOI: 10.1002/anie.201004284] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
23
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemische Kontrolle des Schicksals und Entwicklungspotenzials von Stammzellen. Angew Chem Int Ed Engl 2010. [DOI: 10.1002/ange.201004284] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
24
|
Vaziri H, Chapman KB, Guigova A, Teichroeb J, Lacher MD, Sternberg H, Singec I, Briggs L, Wheeler J, Sampathkumar J, Gonzalez R, Larocca D, Murai J, Snyder E, Andrews WH, Funk WD, West MD. Spontaneous reversal of the developmental aging of normal human cells following transcriptional reprogramming. Regen Med 2010; 5:345-63. [PMID: 20230312 DOI: 10.2217/rme.10.21] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM To determine whether transcriptional reprogramming is capable of reversing the developmental aging of normal human somatic cells to an embryonic state. MATERIALS & METHODS An isogenic system was utilized to facilitate an accurate assessment of the reprogramming of telomere restriction fragment (TRF) length of aged differentiated cells to that of the human embryonic stem (hES) cell line from which they were originally derived. An hES-derived mortal clonal cell strain EN13 was reprogrammed by SOX2, OCT4 and KLF4. The six resulting induced pluripotent stem (iPS) cell lines were surveyed for telomere length, telomerase activity and telomere-related gene expression. In addition, we measured all these parameters in widely-used hES and iPS cell lines and compared the results to those obtained in the six new isogenic iPS cell lines. RESULTS We observed variable but relatively long TRF lengths in three widely studied hES cell lines (16.09-21.1 kb) but markedly shorter TRF lengths (6.4-12.6 kb) in five similarly widely studied iPS cell lines. Transcriptome analysis comparing these hES and iPS cell lines showed modest variation in a small subset of genes implicated in telomere length regulation. However, iPS cell lines consistently showed reduced levels of telomerase activity compared with hES cell lines. In order to verify these results in an isogenic background, we generated six iPS cell clones from the hES-derived cell line EN13. These iPS cell clones showed initial telomere lengths comparable to the parental EN13 cells, had telomerase activity, expressed embryonic stem cell markers and had a telomere-related transcriptome similar to hES cells. Subsequent culture of five out of six lines generally showed telomere shortening to lengths similar to that observed in the widely distributed iPS lines. However, the clone EH3, with relatively high levels of telomerase activity, progressively increased TRF length over 60 days of serial culture back to that of the parental hES cell line. CONCLUSION Prematurely aged (shortened) telomeres appears to be a common feature of iPS cells created by current pluripotency protocols. However, the spontaneous appearance of lines that express sufficient telomerase activity to extend telomere length may allow the reversal of developmental aging in human cells for use in regenerative medicine.
Collapse
Affiliation(s)
- H Vaziri
- Ontario Cancer Institute/PMH, University of Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ilic D. Industry Update: Latest developments in stem cell research and regenerative medicine. Regen Med 2010. [DOI: 10.2217/rme.10.64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Dusko Ilic
- Embryonic Stem Cell Laboratories, Guy’s Assisted Conception Unit, Division of Reproduction & Endocrinology, King’s College London, UK
| |
Collapse
|
26
|
Ilic D. Latest developments in stem cell research and regenerative medicine. Regen Med 2008. [DOI: 10.2217/17460751.3.5.641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Dusko Ilic
- StemLifeLine, Inc., 1300 Industrial Rd #13, San Carlos, CA 94070, USA
| |
Collapse
|