Category Archives: Induced Pluripotent Stem Cells


Challenges and perspectives of heart repair with pluripotent stem cell-derived cardiomyocytes – Nature.com

Nabel, E. G. & Braunwald, E. A tale of coronary artery disease and myocardial infarction. N. Engl. J. Med. 366, 5463 (2012).

Article CAS PubMed Google Scholar

McDonagh, T. A. et al. Group ESCSD. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 42, 35993726 (2021).

Article CAS PubMed Google Scholar

Goldenberg, B. Ueber Atrophie und Hypertrophie der Muskelfasern des Herzens. Arch. Pathol. Anat. Physiol. Klin. Med. 103, 88130 (1886).

Article Google Scholar

Linzbach, A. J. Heart failure from the point of view of quantitative anatomy. Am. J. Cardiol. 5, 370382 (1960).

Article CAS PubMed Google Scholar

Linzbach, A. J. Hypertrophy, hyperplasia and structural dilatation of the human heart. Adv. Cardiol. 18, 114 (1976).

Article CAS PubMed Google Scholar

Soonpaa, M. H., Kim, K. K., Pajak, L., Franklin, M. & Field, L. J. Cardiomyocyte DNA synthesis and binucleation during murine development. Am. J. Physiol. 271, H2183H2189 (1996).

CAS PubMed Google Scholar

Bergmann, O. et al. Evidence for cardiomyocyte renewal in humans. Science 324, 98102 (2009).

Article CAS PubMed PubMed Central Google Scholar

Olivetti, G. et al. Apoptosis in the failing human heart. N. Engl. J. Med. 336, 11311141 (1997).

Article CAS PubMed Google Scholar

Quaini, F. et al. Chimerism of the transplanted heart. N. Engl. J. Med. 346, 515 (2002).

Article PubMed Google Scholar

Orlic, D. et al. Bone marrow cells regenerate infarcted myocardium. Nature 410, 701705 (2001).

Article CAS PubMed Google Scholar

Jeyaraman, M. M. et al. Autologous bone marrow stem cell therapy in patients with st-elevation myocardial infarction: a systematic review and meta-analysis. Can. J. Cardiol. 33, 16111623 (2017).

Article PubMed Google Scholar

Blau, H. M., Brazelton, T. R. & Weimann, J. M. The evolving concept of a stem cell: entity or function? Cell 105, 829841 (2001).

Article CAS PubMed Google Scholar

Dimmeler, S., Zeiher, A. M. & Schneider, M. D. Unchain my heart: the scientific foundations of cardiac repair. J. Clin. Invest. 115, 572583 (2005).

Article CAS PubMed PubMed Central Google Scholar

Beltrami, A. P. et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114, 763776 (2003).

Article CAS PubMed Google Scholar

Oh, H. et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc. Natl Acad. Sci. USA 100, 1231312318 (2003).

Article CAS PubMed PubMed Central Google Scholar

Laugwitz, K. L. et al. Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature. 433, 647653 (2005).

Article CAS PubMed PubMed Central Google Scholar

Planat-Benard, V. et al. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ. Res. 94, 223229 (2004).

Article CAS PubMed Google Scholar

Condorelli, G. et al. Cardiomyocytes induce endothelial cells to trans-differentiate into cardiac muscle: Implications for myocardium regeneration. Proc. Natl Acad. Sci. USA 98, 1073310738 (2001).

Article CAS PubMed PubMed Central Google Scholar

Li, Y. et al. Genetic lineage tracing of nonmyocyte population by dual recombinases. Circulation 138, 793805 (2018).

Article CAS PubMed Google Scholar

Murry, C. E. et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428, 664668 (2004).

Article CAS PubMed Google Scholar

Nygren, J. M. et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat. Med. 10, 494501 (2004).

Article CAS PubMed Google Scholar

Laflamme, M. A., Myerson, D., Saffitz, J. E. & Murry, C. E. Evidence for cardiomyocyte repopulation by extracardiac progenitors in transplanted human hearts. Circ. Res. 90, 634640 (2002).

Article CAS PubMed Google Scholar

Hocht-Zeisberg, E. et al. Cellular repopulation of myocardial infarction in patients with sex-mismatched heart transplantation. Eur. Heart J. 25, 749758 (2004).

Article PubMed Google Scholar

Partners HealthCare and Brigham and Womens Hospital agree to pay $10 million to resolve research fraud allegations. US Attorneys Office District of Massachusetts https://www.justice.gov/usao-ma/pr/partners-healthcare-and-brigham-and-women-s-hospital-agree-pay-10-million-resolve (2017).

Eschenhagen, T. et al. Cardiomyocyte regeneration: a consensus statement. Circulation 136, 680686 (2017).

Article PubMed PubMed Central Google Scholar

Senyo, S. E. et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493, 433436 (2013).

Article CAS PubMed Google Scholar

Vagnozzi, R. J. et al. An acute immune response underlies the benefit of cardiac stem cell therapy. Nature 577, 405409 (2020).

Article CAS PubMed Google Scholar

Sadek, H. & Olson, E. N. Toward the goal of human heart regeneration. Cell Stem Cell 26, 716 (2020).

Article CAS PubMed PubMed Central Google Scholar

Giacca, M. Fulfilling the promise of rna therapies for cardiac repair and regeneration. Stem Cells Transl. Med. 12, 527535 (2023).

Article PubMed PubMed Central Google Scholar

Yamada, Y., Sadahiro, T. & Ieda, M. Development of direct cardiac reprogramming for clinical applications. J. Mol. Cell Cardiol. 178, 18 (2023).

Article CAS PubMed Google Scholar

Soonpaa, M. H., Koh, G. Y., Klug, M. G. & Field, L. J. Formation of nascent intercalated disks between grafted fetal cardiomyocytes and host myocardium. Science 264, 98101 (1994).

Article CAS PubMed Google Scholar

Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 11451147 (1998).

Article CAS PubMed Google Scholar

Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861872 (2007).

Article CAS PubMed Google Scholar

Kehat, I. et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest. 108, 407414 (2001).

Article CAS PubMed PubMed Central Google Scholar

Kattman, S. J. et al. Stage-specific optimization of activin/nodal and bmp signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 8, 228240 (2011).

Article CAS PubMed Google Scholar

Burridge, P. W. & Zambidis, E. T. Highly efficient directed differentiation of human induced pluripotent stem cells into cardiomyocytes. Methods Mol. Biol. 997, 149161 (2013).

Article CAS PubMed Google Scholar

Breckwoldt, K. et al. Differentiation of cardiomyocytes and generation of human engineered heart tissue. Nat. Protoc. 12, 11771197 (2017).

Article CAS PubMed Google Scholar

Halloin, C., Coffee, M., Manstein, F. & Zweigerdt, R. Production of cardiomyocytes from human pluripotent stem cells by bioreactor technologies. Methods Mol. Biol. 1994, 5570 (2019).

Article CAS PubMed Google Scholar

Buikema, J. W. et al. Wnt activation and reduced cellcell contact synergistically induce massive expansion of functional human iPSC-derived cardiomyocytes. Cell Stem Cell 27, 5063 (2020).

Article CAS PubMed PubMed Central Google Scholar

Eschenhagen, T., Ridders, K. & Weinberger, F. How to repair a broken heart with pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol. 163, 106117 (2022).

Article CAS PubMed Google Scholar

Weinberger, F. & Eschenhagen, T. Cardiac regeneration: new hope for an old dream. Annu. Rev. Physiol. 83, 5981 (2021).

Article CAS PubMed Google Scholar

Shiba, Y. et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature 538, 388391 (2016).

Article CAS PubMed Google Scholar

Chong, J. J. et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510, 273277 (2014).

Article CAS PubMed PubMed Central Google Scholar

Liu, Y. W. et al. Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 36, 597605 (2018).

Article CAS PubMed PubMed Central Google Scholar

Querdel, E. et al. Human engineered heart tissue patches remuscularize the injured heart in a dose-dependent manner. Circulation 143, 19912006 (2021).

Article CAS PubMed PubMed Central Google Scholar

Riegler, J. et al. Human engineered heart muscles engraft and survive long term in a rodent myocardial infarction model. Circ. Res. 117, 720730 (2015).

Article CAS PubMed PubMed Central Google Scholar

Laflamme, M. A. et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25, 10151024 (2007).

Article CAS PubMed Google Scholar

Weinberger, F. et al. Cardiac repair in guinea pigs with human engineered heart tissue from induced pluripotent stem cells. Sci. Transl. Med. 8, 363ra148 (2016).

Article PubMed Google Scholar

Bergmann, O. et al. Dynamics of cell generation and turnover in the human heart. Cell 161, 15661575 (2015).

Article CAS PubMed Google Scholar

Lou, X. et al. N-cadherin overexpression enhances the reparative potency of human-induced pluripotent stem cell-derived cardiac myocytes in infarcted mouse hearts. Cardiovasc. Res. 116, 671685 (2020).

Article CAS PubMed Google Scholar

Kobayashi, H. et al. Intracoronary transplantation of pluripotent stem cell-derived cardiomyocytes: inefficient procedure for cardiac regeneration. J. Mol. Cell Cardiol. 174, 7787 (2023).

Here is the original post:
Challenges and perspectives of heart repair with pluripotent stem cell-derived cardiomyocytes - Nature.com

Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications | Signal Transduction and … – Nature.com

Rowe, R. G. & Daley, G. Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 20, 377388 (2019).

Article CAS PubMed PubMed Central Google Scholar

Shi, Y., Inoue, H., Wu, J. C. & Yamanaka, S. Induced pluripotent stem cell technology: a decade of progress. Nat. Rev. Drug Discov. 16, 115130 (2017).

Article CAS PubMed Google Scholar

Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861872 (2007).

Article CAS PubMed Google Scholar

Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663676 (2006).

Article CAS PubMed Google Scholar

Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 19171920 (2007).

Article CAS PubMed Google Scholar

Takahashi, K. & Yamanaka, S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell Biol. 17, 183193 (2016).

Article CAS PubMed Google Scholar

Breschi, A., Gingeras, T. R. & Guigo, R. Comparative transcriptomics in human and mouse. Nat. Rev. Genet. 18, 425440 (2017).

Article CAS PubMed PubMed Central Google Scholar

Gharib, W. H. & Robinson-Rechavi, M. When orthologs diverge between human and mouse. Brief. Bioinform. 12, 436441 (2011).

Article PubMed PubMed Central Google Scholar

Lynch, V. J. Use with caution: developmental systems divergence and potential pitfalls of animal models. Yale J. Biol. Med. 82, 5366 (2009).

CAS PubMed PubMed Central Google Scholar

Takebe, T. & Wells, J. M. Organoids by design. Science 364, 956959 (2019).

Article CAS PubMed PubMed Central Google Scholar

Yamanaka, S. Pluripotent Stem Cell-based Cell Therapy- Promise And Challenges. Cell Stem Cell 27, 523531 (2020).

Article CAS PubMed Google Scholar

Gurdon, J. B. The generation of diversity and pattern in animal development. Cell 68, 185199 (1992).

Article CAS PubMed Google Scholar

Kiefer, J. C. Epigenetics in development. Dev. Dyn. 236, 11441156 (2007).

Article CAS PubMed Google Scholar

Tompkins, J. D. Discovering DNA methylation, the history and future of the writing on DNA. J. Hist. Biol. 55, 865887 (2022).

PubMed PubMed Central Google Scholar

Roe, S. A. Matter, life, and generation: eighteen-century embryology and the Haller-Wolff Debate, (Cambridge University Press, 1981).

Kilgour, F. G. William Harvey and his contributions. Circulation 23, 286296 (1961).

Article CAS PubMed Google Scholar

Aulie, R. P. Caspar Friedrich Wolff and his Theoria generationis, 1759. J. Hist. Med. Allied Sci. 16, 124144 (1961).

Article CAS PubMed Google Scholar

Weismann, A. Das Keimplasma; eine Theorie der Vererbung, (Jena, Fischer, 1892).

Waddington, C. H. The Strategy of the Genes; A Discussion of Some Aspects of Theoretical Biology, (Cambridge: Cambridge University Press, 1957).

Nanney, D. L. Epigenetic control systems. Proc. Natl. Acad. Sci. USA 44, 712717 (1958).

Article CAS PubMed PubMed Central Google Scholar

Gurdon, J. B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 10, 622640 (1962).

CAS PubMed Google Scholar

Gurdon, J. B. The transplantation of nuclei between two species of Xenopus. Dev. Biol. 5, 6883 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B. Adult frogs derived from the nuclei of single somatic cells. Dev. Biol. 4, 256273 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B. Multiple genetically identical frogs. J. Hered. 53, 59 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B., Elsdale, T. R. & Fischberg, M. Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature 182, 6465 (1958).

Article CAS PubMed Google Scholar

Jeltsch, A. & Jurkowska, R. Z. New concepts in DNA methylation. Trends Biochem. Sci. 39, 310318 (2014).

Article CAS PubMed Google Scholar

Riggs, A. D. X inactivation, differentiation, and DNA methylation. Cytogenet. Cell Genet. 14, 925 (1975).

Article CAS PubMed Google Scholar

Robertson, K. D. & Wolffe, A. P. DNA methylation in health and disease. Nat. Rev. Genet. 1, 1119 (2000).

Article CAS PubMed Google Scholar

Schubeler, D. Function and information content of DNA methylation. Nature 517, 321326 (2015).

Article CAS PubMed Google Scholar

Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154156 (1981).

Article CAS PubMed Google Scholar

Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci. USA 78, 76347638 (1981).

Article CAS PubMed PubMed Central Google Scholar

Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 11451147 (1998).

Article CAS PubMed Google Scholar

Tada, M., Takahama, Y., Abe, K., Nakatsuji, N. & Tada, T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr. Biol. 11, 15531558 (2001).

Article CAS PubMed Google Scholar

Cowan, C. A., Atienza, J., Melton, D. A. & Eggan, K. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 309, 13691373 (2005).

Article CAS PubMed Google Scholar

Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51, 9871000 (1987).

Article CAS PubMed Google Scholar

Halder, G., Callaerts, P. & Gehring, W. J. Induction of ectopic eyes by targeted expression of the eyeless gene in drosophila. Science 267, 17881792 (1995).

Article CAS PubMed Google Scholar

Kulessa, H., Frampton, J. & Graf, T. Gata-1 reprograms Avian Myelomonocytic cell-lines into Eosinophils, Thromboblasts, and Erythroblasts. Gene Dev. 9, 12501262 (1995).

Article CAS PubMed Google Scholar

Xie, H., Ye, M., Feng, R. & Graf, T. Stepwise reprogramming of B cells into macrophages. Cell 117, 663676 (2004).

Article CAS PubMed Google Scholar

Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318324 (2007).

Article CAS PubMed Google Scholar

Huangfu, D. W. et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795797 (2008).

Article CAS PubMed PubMed Central Google Scholar

Huangfu, D. et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 26, 12691275 (2008).

Article CAS PubMed Google Scholar

Hou, P. et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341, 651654 (2013).

Article CAS PubMed Google Scholar

Zhu, J. et al. Genome-wide chromatin state transitions associated with developmental and environmental cues. Cell 152, 642654 (2013).

Article CAS PubMed PubMed Central Google Scholar

Apostolou, E. & Hochedlinger, K. Chromatin dynamics during cellular reprogramming. Nature 502, 462471 (2013).

Article CAS PubMed PubMed Central Google Scholar

Apostolou, E. & Stadtfeld, M. Cellular trajectories and molecular mechanisms of iPSC reprogramming. Curr. Opin. Genet. Dev. 52, 7785 (2018).

Article CAS PubMed PubMed Central Google Scholar

Cacchiarelli, D. et al. Integrative analyses of human reprogramming reveal dynamic nature of induced pluripotency. Cell 162, 412424 (2015).

Article CAS PubMed PubMed Central Google Scholar

Nefzger, C. M. et al. Cell type of origin dictates the route to pluripotency. Cell Rep. 21, 26492660 (2017).

Article CAS PubMed Google Scholar

Borkent, M. et al. A serial shRNA screen for roadblocks to reprogramming identifies the protein modifier SUMO2. Stem Cell Rep. 6, 704716 (2016).

Article CAS Google Scholar

Buckley, S. M. et al. Regulation of Pluripotency and cellular reprogramming by the ubiquitin-proteasome system. Cell Stem Cell 11, 783798 (2012).

Article CAS PubMed PubMed Central Google Scholar

Qin, H. et al. Systematic identification of barriers to human iPSC generation. Cell 158, 449461 (2014).

Article CAS PubMed PubMed Central Google Scholar

Simic, M. S. et al. Transient activation of the UPR(ER) is an essential step in the acquisition of pluripotency during reprogramming. Sci. Adv. 5, eaaw0025 (2019).

Article CAS PubMed PubMed Central Google Scholar

Wu, Y. et al. Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition. Sci. Adv. 5, eaax7525 (2019).

Go here to see the original:
Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications | Signal Transduction and ... - Nature.com

Mechanical stimulation of induced pluripotent stem derived cardiac fibroblasts | Scientific Reports – Nature.com

CFs are the main contributors of cardiac fibrosis development3. The availability of human CFs is limited hampering the field to move forward. To date, CFs can be generated from iPSCs, which could provide an unlimited source of human CFs13,14,15. However, the behaviour of iPSC-CFs in relation to mechanical stimulation had not been investigated yet.

In this study we demonstrated that iPSC-CFs are comparable to primary CFs with regard to the expression of key CF markers at gene and protein levels. Expression of the cardiac markers GATA4 and TCF21 indicate the cardiac lineage of the cells. Furthermore, expression of the mesenchymal markers VIM and PDGFRA as well as the ECM component COL1A1 and the collagen binding receptor DDR2 support their fibroblast phenotype. In addition, we showed that iPSC-CFs respond to pro-fibrotic and mechanical stimulation. TGF- induces CFs transdifferentiation into myofibroblasts and promotes ECM remodelling. Mechanical stimulation in the form of cyclic stretch at physiological levels reduces collagen expression in iPSC-CFs. Interestingly, cyclic stretch also protects against TGF- stimulation, preventing the cells from transdifferentiating into myofibroblasts.

One can only use iPSC-derived cells when they accurately represent their primary counterparts. Key characteristics of CFs are a defined mRNA profile, responsiveness to pro-fibrotic cytokines, interaction with the ECM and mechanical sensitivity. iPSC-CFs generated using the protocol developed by Zhang et al. showed a comparable RNA sequencing profile in iPSC-CFs and primary CFs13. Using our iPSCs lines, following the same protocol we generated iPSC-CFs with an mRNA profile comparable to primary CFs. Furthermore, at a functional level we demonstrated that iPSC-CFs interact with their environment in a similar way as primary CFs, and respond to pro-fibrotic stimulation. These results indicate that iPSC-CFs possess several key characteristics of primary CFs and may be suitable to investigate the behaviour of CFs and develop disease models of cardiac fibrosis.

In order to investigate the behaviour of CFs in their native environment, we next investigated the behaviour of iPSC-CFs under physiologically relevant conditions. In an effort to mimic the dynamic environment of the continuously beating heart, we investigated the effects of cyclic mechanical stretch on iPSC-CFs. The importance of mechanical stimulation has been acknowledged, but the effects of mechanical stimulation on CFs remain controversial in in vitro studies23. On one hand, it has been reported that cyclic stretch may induce transdifferentiation of CFs into myofibroblasts. On the other hand, it has been shown that cyclic stretch may have a protective effect instead. One of the main factors influencing this controversy is the usage of cell sources from different species.

As primary human CFs are limited in availability, iPSC-CFs could provide a representative and stable source of cells to move forward. In order to study how iPSC-CFs and primary CFs behave in a mechanically dynamic environment similar to the heart, cells were exposed to 10% cyclic stretch at 1Hz for 72h19. With this approach, we demonstrated that: Cyclic stretch alone inhibits expression of collagen 1 but does not affect iPSC-CFs transdifferentiation or expression of matrix remodelling genes. In addition, cyclic stretch is protective against TGF- mediated myofibroblast transdifferentiation in iPSC-CFs, resulting in normalised expression of collagen 1, -SMA and matrix remodelling genes such as TIMP1 and MMP1.

The cause of the aforementioned controversy in literature regarding either the pro-fibrotic or anti-fibrotic response of CFs to mechanical stimulation is hard to pin-point; experimental conditions vary widely between studies, such as cell origin, the duration of the experiment, the surface coating and the presence of serum. A common trend in all those studies is that there may be a time-dependent response of stretch. It was shown in primary mouse CFs that the response starts with an initial increase in phosphorylation of AKT, a downstream kinase involved in the transduction of mechanical stimuli24,25. At the gene level, it was shown in primary rat CFs that there is an initial increase in fibrotic markers (i.e. ACTA2, TGFB1, CTGF) after 4 h followed by a reduced increase after 24h26. Roche et al. observed a similar effect in primary rat CFs with an apparent reduced increase of COL1A1 gene expression after 48h compared to 24h27. 72h of cyclic stretch was instead shown to inhibit TGF- induced fibroblast activation in primary human CFs16,18. Furthermore, it has been demonstrated that 96h of cyclic stretch can promote or inhibit the response of primary mouse CFs to a broad spectrum of biochemical stimuli, including TGF-, angiotensin II, interleukin-1 and others17. Overall, it appears that longer stimulation results in a gradual decrease of an initial pro-fibrotic response with eventually cells balancing the fibrotic response to the mechanically active environment in order to reach homeostasis. We may hypothesize that the duration of this response curve is dependent on different factors, including the origin and age of the cells, their culture conditions (surface coating, substrate stiffness, or medium supplementation with serum) and the presence of other cell types23. A clear association between mechanosensing and a response of CFs is apparent, but there is a need for a reproducible cell type to better understand this phenomenon.

TGF- signalling is one of the main pathways involved in the activation of CFs and development of cardiac fibrosis28. Exposure of iPSC-CFs to TGF- promotes the expression of fibrotic and myofibroblast markers, such as -SMA. When stretched however, this effect is diminished. How mechanical changes communicate with the TGF- pathway is not well understood. On one hand, mechanical strain has been shown in tissue to release active TGF- from the ECM, which would promote fibroblast activation29. On the other hand, in this in vitro study mechanical strain appears to inhibit fibroblast activation, indicating that there may be other mechanisms at play in this model. It is unknown whether this anti-fibrotic effect is directly caused by interplay between mechanosensitive complexes and the TGF- pathway. Mechanosensitive receptors such as integrins or mechanoresponsive factors such as YAP/TAZ may communicate with the TGF- pathway30,31. Alternatively, cyclic stretch may have an indirect effect, for example through internalization of extracellular receptors, altering the response to ligand stimulation. Regardless, the field of mechanotransduction in CFs remains requires further investigation.

While iPSCs have started a new era of research, the usage of these cells comes with limitations. iPSC-CFs showed many similarities with primary CFs, but the maturity of iPSC-derived cell lineages remains an important topic of contention. Although maturation is clearly defined for some cell types, such as cardiac myocytes, a clear definition lacks for CFs. The heterogeneity and plasticity of this cellular population under physiological conditions makes it difficult to set well defined standards of mature CFs32. iPSC-CFs present with various characteristics of primary cells, but they differ in several aspects as well. For example, Zhang et al. noted an increased proliferation capacity in iPSC-CFs and foetal CFs compared to adult CFs, indicating the iPSC-CFs may be more foetal-like13. This increased proliferation capacity and ability to stay in an inactivated state while in culture increases the applicability of the iPSC-CFs in research, as it has been demonstrated that CFs which have transdifferentiated into myofibroblast will have an altered response to mechanical stimulation33. In addition, little is known about the electrophysiological characteristics of iPSC-CFs and their interaction with other conducting cells such as cardiomyocytes34. Further electrophysiological characterisation should be performed to better understand the behaviour of these cell in the electrical circuit of the heart.

To conclude, in this study we demonstrated that iPSC-derived CFs show similar gene and protein expression as primary CFs. In addition, pro-fibrotic stimulation promoted transdifferentiation of iPSC-CFs into a myofibroblast phenotype. When stimulated with cyclic stretch, this transdifferentiation is inhibited. Together, the mechano- and TGF--responsive characteristics support the use of iPSC-CFs for physiological relevant disease modelling. Future studies could further dive into the mechanisms driving cardiac fibroblast behaviour and cardiac fibrosis.

Read more from the original source:
Mechanical stimulation of induced pluripotent stem derived cardiac fibroblasts | Scientific Reports - Nature.com

Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia … – Nature.com

Cell culture

Eight human iPSC lines were employed in this study (Supplementary TableS1). The cells were cultured using Corning Matrigel hESC-Qualified Matrix (Corning, Cat. No. 354277) coated plates with the use of StemMACS iPS Brew XF Medium (Miltenyi Biotec, Cat. No. 130-104-368) or Essential 8 medium (ThermoFisher Scientific, Cat. No. A157001), in antibiotic-free conditions, and maintained at 37C, 5% CO2. iPSCs were passaged every 35 days using either Accutase or 0.5mM phosphate-buffered saline (PBS)/EDTA. Briefly, when passaging the cells with Accutase, cells were firstly washed with DMEM, 1ml of Accutase was added per 6-well and the cells were incubated at 37C for 34min, to ensure proper cell detachment. After the incubation an equal volume of DMEM was added to the well and the cells were collected and centrifuged at 1200rpm for 3min at 4C. For splitting with PBS/EDTA (ThermoFisher Scientific, Cat. No. 15575020), cells were briefly washed with DMEM, 1ml PBS/EDTA was added per 6-well and the cells were incubated until they started to roughly dissociate. The EDTA was aspirated and the cells or the cell pellet (when splitting using Accutase), were resuspended in fresh medium supplemented with 10M ROCK inhibitor, Y-27632, (Miltenyi Biotec, Cat. No. 130-106-538). The next day the medium was changed back to iPSC medium without ROCK inhibitor. All cell lines were thoroughly characterized for their pluripotency (Supplementary Fig.1A, B) and were tested frequently for mycoplasma contamination.

The generation of cortical progenitors and neurons was performed as described before [27, 28] with minor modifications. Briefly, iPSCs from five 6-wells were collected with Accutase and seeded onto an ES-Matrigel coated 12-well. When 100% confluency was reached, StemMACS iPS Brew XF Medium was replaced by neural induction medium (NIM; DMEM/F12 Glutamax, Neurobasal, 100 mM L-Glutamine, 0.5N-2, 0.5B-27+Vitamin A, 50M Non-Essential Amino Acids, 50M 2-mercaptoethanol, 2.5g/ml insulin, 1M dorsomorphine, 10M SB431542). The medium was changed every day until the appearance of a tightly packed neuroepithelial sheet (NES). NES was passaged with 0.5mM EDTA in a ratio of 1:2 or 1:3 to Corning Matrigel Growth Factor Reduced (GFR) Basement Membrane Matrix (GFR-Matrigel; Corning, Cat. No. 354230) coated plates. The next day, the medium was switched to neural maintenance medium (NMM; DMEM/F12 Glutamax, Neurobasal, 100mM L-Glutamine, 0.5N-2, 0.5B-27+Vitamin A, 50M Non-Essential Amino Acids, 50M 2-mercaptoethanol, 2.5g/ml insulin) and was changed every other day. Upon the appearance of rosettes, 20ng/ml FGF2 (Peprotech, Cat. No. 100-18C) were added to the medium for four days. On the fourth day of FGF2 treatment, the cells were split again with 0.5mM EDTA in a ratio of 1:2 to 1:3 onto GFR-Matrigel coated plates. The medium was switched back to NMM and the cortical progenitors were maintained for about 510 days until neurons accumulated outside of the rosettes. At this point, cells were passaged with Accutase, and 50,000 cells/cm were seeded on poly-L-ornithin/Laminin coated plates for further neuronal differentiation. Alternatively, 24million cells/ml were frozen with neural freezing medium. Neurons differentiated further with half medium changes every two to three days. Samples were harvested at day (d) 0, 7, 16, 27, 50, and 100.

For the DFMO treatment, adherent cell cultures were treated daily with 10M DFMO (difluoromethylornithine hydrochloride hydrate; Merck, Cat. No. D193) starting from the first day of differentiation until the collection of cellular pellet and supernatant for mass spectrometry analysis or fixation for subsequent immunocytochemistry (ICC).

Cells were fixed in 4% paraformaldehyde (PFA; Sigma, Cat. No. 158127-500G) in PBS solution for 20min at room temperature (RT). The non-specific binding was blocked with incubation with blocking buffer (3% bovine serum albumin (BSA), 0.2% Triton 100 in PBS) for 1h at RT. The primary antibody (Ab) was diluted in the blocking buffer in the recommended concentration and the Ab solution was applied overnight at 4C. The following primary Abs were used in the following concentrations: AFP 1:400 (Dako, Cat. No. A000829-2), GAD65/67 1:100 (Abcam, Cat. No. AB183999), GFAP 1:400 (Sigma, Cat. No. G3893-.2ML), Ki67-VioR667 1:200 (Miltenyi, Cat. No.130-120-422), MAP2 1:1,000 (SynapticSystems, Cat. No.188006), NEUN 1:500 (Sigma, Cat. No. ABN78), OCT3/4 1:200 (Szabo-Scandic, Cat. No. GTX101497-100), PAX6 1:500 (Invitrogen, Cat. No. 42-6600), S100b 1:750 (Abcam, Cat. No. ab52642), SMA 1:500 (Abcam, Cat. No. ab7817), SOX1 1:200 (R&D Systems, Cat. No. AF3369), SOX2 1:500 (R&D Systems, Cat. No. MAB2018), TAU 1:200 (Cell Signaling Technology, Cat. No. 4019), TUBB3 1:1,000 (BioLegend, Cat. No. 801202 and Abcam, Cat.No. ab52623), vGLUT 1:100 (SynapticSystems, Cat. No. 135311). The secondary Ab was diluted 1:500 in 1.5% BSA, 0,2% Triton 100 in PBS, and the solution was applied for 2h at RT. The secondary Abs used in this study were: donkey anti-rabbit Alexa FluorTM 488 (ThermoFisher Scientific, Cat. No. A-21206), donkey anti-rabbit Alexa FluorTM 546 (ThermoFisher Scientific, Cat. No. A-10040), donkey anti-mouse Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A-21203), donkey anti-mouse Alexa FluorTM 647 (ThermoFisher Scientific, Cat. No. A-31571), donkey anti-goat Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A-11058), goat anti-chicken Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A32759). Finally, the nuclei were counterstained using 4,6-diamidino-2-phenylindole (DAPI; ThermoFisher Scientific, Cat. No. D21490) in PBS in 1:5000 dilution for 5min at RT. The coverslips were mounted using Aqua-Poly/Mount mounting medium (PolySciences, Cat. No. 18606-20).

Fluorescent pictures were acquired with the Zeiss Axio Observer Z1 inverted fluorescent microscope and the Leica DMi8 inverted microscope. The image acquisition was performed under the same exposure and laser intensity settings for each set of analyses. For each sample, ten random fields of view were acquired, with a minimum of 20 z-stacks collected per field to ensure proper signal coverage. Further image processing was carried out using the ImageJ software. For quantitative fluorescence intensity analysis, maximum intensity projection was applied and the mean fluorescence intensity values were calculated after background noise subtraction. These values were then normalized to the DAPI+ nuclear area to account for variations in cell density in the different fields of view.

Total RNA was extracted from cells using TRI Reagent (Merck, Cat. No. T9424), according to the manufacturers instructions. Genomic DNA was removed through treatment with DNase I (Sigma-Aldrich, Cat. No. AMPD1). Subsequently, 1g of purified RNA was reverse transcribed into cDNA using the RevertAid RT Reverse Transcription Kit (ThermoFisher Scientific, Cat. No. K1691), following the manufacturers guidelines. The expression levels of specific target genes at the mRNA level were quantified via reverse transcription quantitative PCR (RT-qPCR) using the 5 HOT FIREPol EvaGreen qPCR Mix Plus (no ROX) (Solis BioDyne, Cat. No. 08-25-00001-10). Samples were analyzed in technical triplicates to ensure data reliability. Non-template controls (NTCs) were included for each primer pair in every assay to monitor for reagent contamination and primer-dimer formation. To confirm the absence of genomic DNA contamination, random RNA samples were evaluated through gel electrophoresis. The RT-qPCR assays were conducted on the CFX Connect Real-Time PCR Detection System (Bio-Rad). Gene expression levels were normalized to the housekeeping gene ACTB. Relative expression changes were calculated employing the Ct method [29]. The list of the primers used for RT-qPCR assays is shown in Table1.

Total RNA was isolated from cells at six time points during the cortical differentiation and was prepared for paired-end mRNA sequencing. RNA extraction was performed using the TRI Reagent (Merck, Cat. No. T9424) according to the manufacturers guidelines. Genomic DNA digest was performed with the use of the TURBO DNA-free Kit (ThermoFisher Scientific, Cat. No. AM2238). For the library preparation, the Illumina TruSeq RNA Library Prep Kit v2 was used (Illumina, Cat. No. RS-122-2001, RS-122-2002). Quality, as well as concentration of RNA were assessed employing the Agilent RNA 6000 Pico kit (Agilent, cat. no. 5067-1513), Nanodrop, the NEBNext Library Quant Kit for Illumina (New England Biolabs, Cat. No. E7630S) and the Qubit RNA Integrity and Quality (IQ) Assay Kit (ThermoFisher Scientific, Cat. No. Q33222). All the kits were used according to the manufacturers guidelines. Paired-end sequencing was performed with the NextSeq 500/550 v2 Kit (150 cycles) (Illumina).

Low-quality ends and adapter sequences were trimmed using the wrapper Trim Galore!. Reads were mapped to the human reference genome (GRCh38) using the open-source software STAR [30]. The raw counts were generated with the Hypergeometric Optimization of Motif EnRichment (HOMER) suite [31]. All the subsequent analysis was performed using R [32]. Differential gene expression analysis was performed using the DESeq2 package [33]. Raw counts were normalized using the median of ratios (variance stabilization transformation; vst) [34]. Heatmaps were generated with the ClustVis [35] tool, using the z-score of the vst transcriptomic data for every gene. Gene ontology (GO) enrichment analysis was performed using the ShinyGO 0.76 online tool [36].

A likelihood ratio test (LRT) was used to identify the differentially expressed genes (DEGs) of SCZ and control (CTRL) across the multiple time points of neuronal differentiation [32]. The LRT compared the full model containing the covariates sex, batch, time point, and disease with a model reducing the covariates sex, batch, and time point. Statistical values were corrected for FDR using the Benjamini-Hochberg method.

Weighted Gene Correlation Network Analysis (WGCNA) allows the generation of modules that include genes that are co-expressed in the same manner. The vst counts were used to build a co-expression network using the WGCNA [37] package in R [32]. The data were corrected for sex and batch effects using the ComBat function that is implemented in the sva package [38]. The topological overlap measure was calculated using the adjacency matrix. The DynamicTree Cut algorithm, implemented in the WGCNA package, was used to identify the different modules. The gray module contains all the genes that were not assigned to any of the other modules. The module eigengene were calculated. Pearsons correlation was used to compare modules to each other and to the traits SCZ and the differentiation time points in the adjacency matrix. The top 25% of genes with the highest module membership (MM) were identified as hub genes.

Functional enrichment analysis was performed with an input gene ID list using the tool g:GOSt from the g:Profiler [39] R package. Statistical significance was computed and the g:SCS-threshold was corrected at p<0.05.

The cells were washed with 1ml sterile 1x PBS for 60s. After the wash, the cells were scraped using 1ml PBS and the suspension was collected and centrifuged at 4000rpm for 5min, at RT. The cell pellets were kept constantly on dry ice and stored at 80C until further processing. The cell supernatant was collected after a 24-h incubation, centrifuged at 4000rpm for 10min, immediately placed on dry ice and stored at 80C. Samples were analyzed using the biocrates MxP Quant 500 (biocrates life sciences AG, Cat. No. 21094.12). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was employed to analyze small molecules, including analyte classes such as amino acids, biogenic amines, carboxylic acids, and amino acid-related molecules [40]. Lipid species were measured using flow injection analysis tandem mass spectrometry (FIAMS/MS). Small molecules were quantified with external 7-point calibrations and internal standards and lipids were quantified by internal standards [41]. The raw data were processed by applying a modified 80% rule to reduce the false positive measurements [42]. The actual missing values, i.e., the values over the level of detection (LOD) for one time point but not for another time point, were uniformly at random imputed with a non-zero value between LOD/2 and LOD. Missing values within one class(i.e., timepoints and metabolites) were imputed using the arithmetic mean of the class. Batch effects were corrected by centering the data within the groups(i.e., time points) and batches. The performance of the normalization was assessed by plotting the row standard deviations versus the row means and the principal component analysis (PCA). In addition, variancePartition analysis was performed to evaluate the contribution of each individual component of the study design (i.e., time point, batch, and condition), to the measured variation of each metabolite [43].

For metabolite extraction, cell pellets were resuspended in 500L ice-cold methanol. Metabolites from supernatants (50L) were extracted using 450L 8:1 methanol:water. Fully 13C, 15N labeled amino acid standard (Cambridge Isotope Laboratories, Cat. No. MSK-CAA-1) and 6D-gamma hydroxybutyrate (Sigma-Aldrich, Cat. No. 615587) were spiked into samples at the first step of the extraction. After simultaneous proteo-metabolome liquid-liquid extraction [44], protein content was determined from extracted cellular interphases using a Pierce Micro BCA Protein Assay Kit (Thermo Fisher Scientific, Cat. No. 23235). Dried metabolite samples from cell pellets were dissolved in 20L 0.1% formic acid (FA) or 50L 0.1% FA for the analysis from the supernatant samples. The sample (1L) was injected on an Atlantis Premier BEH C18 AX column (1.7m, 2.1150mm, Waters, 186009361) equilibrated at 40C using an Acquity Premier UPLC system (Waters). A gradient was run at a flowrate of 0.4mL/min with mobile phase A (0.1% FA in water) and mobile phase B (0.1% FA in acetonitrile) as follows: 1min at 1% B, to 40% B in 1min, 40% B to 99% B in 0.5min, hold at 99% B for 1.1min, 99% B to 1% B in 0.1min followed by 1.8min of re-equilibration at 1% B. GABA and Glutamate (Glu) were detected using a Xevo-TQ XS Mass spectrometer (Waters) equipped with an electrospray ionization source running in positive mode. The transitions 104>69 (endogenous GABA), 110>73 (labeled GABA), 148>102 (endogenous Glu) and 154>107 (labeled Glu) were used for quantification. The raw files were processed using MS Quan in waters connect (Waters, V1.7.0.7). The data was further analyzed in R and normalized to the protein content.

To analyze time-related cluster dynamics, the non-parametric clustering algorithm of Short Time-series Expression Miner (STEM) was used [45]. STEM is an online tool that assigns genes or metabolites to significant temporal expression profiles. The Maximum Number of Model Profiles and the Maximum Unit Change in Model Profiles between time points were set to 50 and 2, respectively. Data were normalized to d0. Integrated into the STEM tool is a GO enrichment analysis. All annotations (Biological Process (BP), Molecular Function (MF), and Cellular Component (CC)) were selected and applied. Statistical significance was computed and FDR-corrected at p<0.05.

The network establishment was based on the gene expression and metabolite level changes across the five successive time point comparisons, along the cortical differentiation. The connectivity information for the initial network was acquired from the publicly available recon3D stoichiometric model data set (available at https://www.vmh.life/#downloadview, retrieved in September 2020) [46]. Ultimately, 51 metabolites and 1135 genes were matched with their corresponding IDs.

Briefly, the construction of the network was performed based on the following steps. Initially, all the reactions associated with any of the target genes were extracted. The metabolites associated with these reactions were identified and the educt-product stoichiometry was applied for every metabolite involved in the network. Subsequently, the reaction data were filtered to extract and proceed only with the genes and metabolites measured in our dataset. The network was further enriched with protein-protein interaction information, derived using the signor database (available at https://signor.uniroma2.it/downloads.php, retrieved in September 2020) [47]. Finally, the network vertices were constructed after examining the unique metabolites and genes, existing in the edge dataset and were further enriched with vertex attributes, such as the vertex type (i.e., gene/metabolite). Log2 fold changes (log2FC) were converted to a color gradient scale, ranging from blue (indicating a downregulation compared to the previous time point) to red (indicating upregulation).

Extraction of subnetworks from the parental network, was based on assigning membership to the pathways, as defined by the KEGG pathway database, and selecting the subnetwork that included the highest number of differentially expressed genes and metabolites, with the closest degree distribution of the vertices. Pie charts with five equal fractions were used in order to visualize the fold changes occurring across a single metabolite or gene, corresponding to the transitions between two succeeding time points. Additionally, ellipses were used for visualizing the metabolites, while the genes were visualized with circles.

Metabolites that were needed as substantial interconnections between measured metabolites, but were not measured in our dataset, were visualized as small dots. The position for every node was provided as coordinates on a 2D plane. Network visualizations were performed using the R igraph package [48].

Follow this link:
Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia ... - Nature.com

Accelerating cardiac regenerative therapy with HiPSC spheroids – Drug Target Review

Injections of cardiac spheroids into primate ventricles improved left ventricular ejection after four weeks.

Researchers from Shinshu University and Keio University School of Medicine have tested a novel strategy for regenerative heart therapy. They transplanted cardiac spheroids derived from human induced pluripotent stem cells (HiPSCs) into damaged ventricles and observed very positive outcomes in primate models. These results could expand treatment options for people suffering from heart problems.

The prevalence of myocardial infarction is rising. These destroy millions of cardiac muscle cells, leaving the heart in a weakened state. Currently, as mammals cannot regenerate cardiac muscle cells on their own, heart transplants are the only clinically viable option for patients suffering heart failure. However, full heart transplants are expensive and donors are rare, so alternative therapies are highly sought after.

The team cultivated HiPSCs in a medium that led to their differentiation into cardiomyocytes. Following the extraction and purification of cardiac spheroids, they injected approximately 6 107cells into the damaged hearts of crab-eating macaques and monitored the condition of the animals for twelve weeks, taking regular measurements of cardiac function.

Analysis of the monkeys hearts at the tissue level was then conducted to assess whether cardiac spheroids could regenerate the damaged heart muscles. The researchers verified the correct reprogramming of HiPSCs into cardiomyocytes first, observing at cellular-level electrical measurements that the cultured cells showed patterns typical of ventricular cells. Also, the cells responded as expected to numerous known drugs. Significantly, they discovered that the cells abundantly expressed adhesive proteins like connexin 43 and N-cadherin, which would promote their vascular integration into an existing heart.

Furthermore, this approach is less expensive and easier to adopt because the cells were transported from the production facility at Keio University to Shinshu University, located 230km away. The cardiac spheroids were preserved at 4C in standard containers and withstood the four-hour journey, meaning extreme cryogenic measures would not be required when transporting the cells to clinics.

The monkeys received injections of either cardiac spheroids or a placebo directly into the damaged heart ventricle. The team noted that arrhythmias were very uncommon, with only two individuals experiencing transient tachycardia in the first two weeks among the treatment group. Echocardiography and computed tomography exams confirmed that, compared to the control group, the hearts of monkeys that received treatment had better left ventricular ejection after four weeks, demonstrating a superior blood pumping capability.

Ultimately, it was revealed through the histological analysis that the cardiac grafts were mature and properly connected to pre-existing existing tissue, confirming the results of previous observations. HiPSC-derived cardiac spheroids could potentially serve as an optimal form of cardiomyocyte products for heart regeneration, given their straightforward generation process and effectiveness, explained first author Dr Hideki Kobayashi. We believe that the results of this research will help solve the major issue of ventricular arrhythmia that occurs after cell transplantation and will greatly accelerate the realisation of cardiac regenerative therapy.

Despite this cardiac spheroid production protocol being tested in monkeys, it was designed for clinical application in humans. The favourable results obtained thus far are sufficient to provide a green light for our clinical trial, called the LAPiS trial. We are already employing the same cardiac spheroids on patients with ischemic cardiomyopathy, concluded Dr Kobayashi.

This study was published in Circulation.

More here:
Accelerating cardiac regenerative therapy with HiPSC spheroids - Drug Target Review

Using stem cell-derived heart muscle cells to advance heart regenerative therapy – EurekAlert

image:

Study shows that cardiac spheroids, derived from human induced pluripotent stem cells, can be easily transported and injected into damaged areas of the heart to promote its regeneration and recovery of function.

Credit: Hideki Kobayashi from Shinshu University

Regenerative heart therapies involve transplanting cardiac muscle cells into damaged areas of the heart to recover lost function. However, the risk of arrhythmias following this procedure is reportedly high. In a recent study, researchers from Japan tested a novel approach that involves injecting cardiac spheroids, cultured from human stem cells, directly into damaged ventricles. The highly positive outcomes observed in primate models highlight the potential of this strategy.

Cardiovascular diseases are still among the top causes of death worldwide, and especially prevalent in developed countries. Myocardial infarctions, commonly known as heart attacks, are on the rise, resulting in a significant number of deaths each year.

Heart attacks typically kill millions of cardiac muscle cells, leaving the heart in a weakened state. Since mammals cannot regenerate cardiac muscle cells on their own, heart transplants are currently the only clinically viable option for patients suffering (or likely to suffer) heart failure. Given that full heart transplants are expensive and donors difficult to come by, it is no surprise that alternative therapies are highly sought after by the medical community.

One promising strategy that has been steadily gaining traction is using human induced pluripotent stem cells (HiPSCs) for regenerative heart therapy. Simply put, HiPSCs are cells derived from mature cells that can be effectively reprogrammed into a completely different cell type, such as cardiac muscle cells (cardiomyocytes). By transplanting or injecting cardiomyocytes derived from HiPSCs into damaged areas of the heart, it is possible to recover some lost functionality. Unfortunately, studies have reported that this approach can increase the risk of arrythmias, posing a major hurdle to clinical trials.

In a recent study, a Japanese research team from Shinshu University and Keio University School of Medicine, tested a new strategy for regenerative heart therapy that involves injecting cardiac spheroids derived from HiPSCs into monkeys with myocardial infarction. This study, published on April 26, 2024, in the journal Circulation, was led by Professor Yuji Shiba from the Department of Regenerative Science and Medicine, Shinshu University.

The team included Hideki Kobayashi, the first author, and Koichiro Kuwahara from the Department of Cardiovascular Medicine, Shinshu University School of Medicine, as well as Shugo Tohyama, and Keiichi Fukuda from the Department of Cardiology, Keio University School of Medicine, among others.

In their novel approach, the researchers cultivated HiPSCs in a medium that led to their differentiation into cardiomyocytes. After carefully extracting and purifying cardiac spheroids (three-dimensional clusters of cardiac cells) from the cultures, they injected approximately 6 107 cells into the damaged hearts of crab-eating macaques (Macaca fascicularis). They monitored the condition of the animals for twelve weeks, taking regular measurements of cardiac function. Following this, they analyzed the monkeys hearts at the tissue level to assess whether cardiac spheroids could regenerate the damaged heart muscles.

First, the team verified the correct reprogramming of HiPSCs into cardiomyocytes. They observed, via cellular-level electrical measurements, that the cultured cells exhibited potential patterns typical of ventricular cells. The cells also responded as expected to various known drugs. Most importantly, they found that the cells abundantly expressed adhesive proteins such as connexin 43 and N-cadherin, which would promote their vascular integration into an existing heart.

Afterwards, the cells were transported from the production facility at Keio University to Shinshu University, located 230 km away. The cardiac spheroids, which were preserved at 4 C in standard containers, withstood the four-hour journey without problem. This means that no extreme cryogenic measures would be needed when transporting the cells to clinics, which would make the proposed approach less expensive and easier to adopt.

Finally, the monkeys received injections of either cardiac spheroids or a placebo directly into the damaged heart ventricle. During the observation period, the researchers noted that arrythmias were very uncommon, with only two individuals experiencing transient tachycardia (fast pulse) in the first two weeks among the treatment group. Through echocardiography and computed tomography exams, the team confirmed that the hearts of monkeys that received treatment had better left ventricular ejection after four weeks compared to the control group, indicating a superior blood pumping capability.

Histological analysis ultimately revealed that the cardiac grafts were mature and properly connected to pre-existing existing tissue, cementing the results of previous observations. HiPSC-derived cardiac spheroids could potentially serve as an optimal form of cardiomyocyte products for heart regeneration, given their straightforward generation process and effectiveness, remarks Assistant Professor Kobayashi. We believe that the results of this research will help solve the major issue of ventricular arrhythmia that occurs after cell transplantation and will greatly accelerate the realization of cardiac regenerative therapy, he further adds.

Although tested in monkeys, it is worth noting that the cardiac spheroid production protocol used in this study was designed for clinical application in humans. The favorable results obtained thus far are sufficient to provide a green light for our clinical trial, called the LAPiS trial. We are already employing the same cardiac spheroids on patients with ischemic cardiomyopathy, comments Asst. Prof. Kobayashi.

Let us all hope for a resounding success in the LAPiS trial, paving the way for expanded and effective treatment avenues for people suffering from heart problems.

###

About Shinshu University

Shinshu Universityis a national university founded in 1949 and located nestling under the Japanese Alps in Nagano known for its stunning natural landscapes. Our motto, "Powered by Nature - strengthening our network with society and applying nature to create innovative solutions for a better tomorrow" reflects the mission of fostering promising creative professionals and deepening the collaborative relationship with local communities, which leads to our contribution to regional development by innovation in various fields. Were working on providing solutions for building a sustainable society through interdisciplinary research fields: material science (carbon, fiber and composites), biomedical science (for intractable diseases and preventive medicine) and mountain science, and aiming to boost research and innovation capability through collaborative projects with distinguished researchers from the world. For more information visit https://www.shinshu-u.ac.jp/english/ or follow us on X (Twitter) @ShinshuUni for our latest news.

About Assistant Professor Hideki Kobayashi

Prof. Hideki Kobayashi became a faculty member at Shinshu University Graduate School of Medicine in 2017, focusing his expertise in cardiology and cardiac electrophysiology. With a prolific publication record, he has contributed to over 25 papers in these fields. His professional affiliations include membership in esteemed organizations such as the Japan Society of Internal Medicine, the Japan Circulation Society, the Japanese Heart Rhythm Society and the International Society of Cardiology Research.

About Professor Yuji Shiba

Prof. Yuji Shiba obtained MD and PhD degrees from Shinshu University in 1998 and 2007, respectively, and has remained closely affiliated with the institution throughout his career. Since 2017, he has been a full Professor at the Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research at Shinshu University. His research endeavours primarily focus on cardiac regeneration using stem cells, an emerging biotechnological field. He has published over 45 papers on these topics, as well as over 10 books and book chapters. He is a member of the Japan Society of Internal Medicine and the Japan Circulation Society.

Experimental study

Animals

Regeneration of non-human primate hearts with human induced pluripotent stem cell-derived cardiac spheroids

26-Apr-2024

The authors have no competing interests to declare.

Continue reading here:
Using stem cell-derived heart muscle cells to advance heart regenerative therapy - EurekAlert

Investigating the promising future of organoids – Drug Target Review

In this podcast episode, titled Investigating the promising future of organoids, we will be discussing the ways in which organoids are being used to study diseases and improve drug development. We explore the specific preclinical models organoids are replacing, the current limitations of organoids and how to overcome these, as well as where the field is heading.

This conversation features Dr Radhika Menon, Senior Scientist of Neurobiology at Ncardia,Dr Etienne De Braekeleer, Senior Research Scientist at AstraZeneca, and Dr Paige Vinson, Director of High Throughput Screening at Southern Research.

This podcast is in association with Molecular Devices. With its innovative life science technology, Molecular Devices makes scientific breakthroughs possible for academic, pharmaceutical, government and biotech customers. Head to moleculardevices.comto find out more.

About the speakers

Dr Radhika Menon, Senior Scientist of Neurobiology at Ncardia

Radhika Menon, PhD is a Senior Scientist at Ncardia, Leiden. In her current position, she leads projects involving human induced pluripotent stem cells (iPSCs) and various iPSC-derived cell types. She started her scientific career as a Junior Researcher in India where she worked on human iPSCs and (embryonic stem cells) ESCs and a cellular model for Alzheimers Disease. She received her PhD in Translational Neuroscience from the Johannes Gutenberg University, Mainz, Germany, working under the supervision of Professor Marisa Karow and Professor Benedikt Berninger. Her PhD project involved the establishment and characterization of an iPSC-derived brain organoid model to study a neurodevelopmental disorder (Opitz Syndrome). Her motivation to do impactful work in the fields of disease modelling, drug development and patient care guided her move to industry to pursue translational research. She moved to the Netherlands to join Mimetas, an organ-on-a-chip company, where she worked with human neuronal, cancer and intestinal biology systems. Dr Menons expertise in cellular differentiation (2D & 3D) and disease modelling in various therapeutic areas is leveraged at Ncardia for cell manufacture and model development services.

Dr Paige Vinson, Director of High Throughput Screening at Southern Research

Paige Vinson, PhD leads a team of scientists in the Southern Research High-Throughput Screening Center in Birmingham, Alabama, where she serves as Director. She earned her PhD in Analytical Chemistry with a focus in Neuroscience from Emory University in Atlanta, Georgia followed by postdoctoral training in Biochemistry at Emory.

Early in her professional career, she gained experience in providing high-throughput drug discovery solutions to researchers in both academia and industry as part of the laboratory automation business unit at Thermo Fisher Scientific. Following this role, Vinson held a research faculty position at Vanderbilt University where she spent twelve years participating in team science, including drug discovery efforts, both as director of HTS and in the molecular pharmacology group of the Warren Center for Neuroscience Drug Discovery.

An ongoing theme in Vinsons career is the bridging of basic research into a drug discovery space through novel approaches in assay development, implementation, and automation including more challenging translational in vitro models. In her current role at Southern Research, Vinson enjoys combining her passions for data-driven approaches and team science to advance projects toward the clinic.

Dr Etienne De Braekeleer, Senior Research Scientist at AstraZeneca

Read more:
Investigating the promising future of organoids - Drug Target Review

HOIL-1L deficiency induces cell cycle alteration which causes immaturity of skeletal muscle and cardiomyocytes … – Nature.com

hiPSC culture

hiPSCs were generated from an Asian female HOIL-1L-deficient patient and healthy controls and kindly donated by the Center for iPS Cell Research and Application (Kyoto University, Kyoto, Japan). Patient-specific (HOIL-1L_1, CiRA-j-0154B and HOIL-1L_2, CiRA-j-0154D) and healthy control hiPSCs (Control_1, CiRA-j-1616-A, Asian female volunteer) were established from the peripheral blood mononuclear cells (PBMCs) using episomal vectors containing reprograming factors30. Another control hiPSC line from Asian male (Control_2, 110F5) was established as described previously31. Each cell line stored in liquid nitrogen using STEM-CELLBANKER (Takara, Cat.# 11924) and once thawed in 37C water bath, it was maintained in mTeSR1 medium (Stem Technologies, Cat.# 85,850) as previously reported32. Each cell was stocked at less than 15 passages, and all experimentations were done between 20 and 45 passages. The sequence of RBCK1 gene was confirmed by Sanger sequencing at the beginning of key experiments. Pluripotency of CiRA-j-1616-A, CiRA-j-0154B and CiRA-j-0154D was evaluated by OCT3/4 and NANOG mRNA expression by TaqMan qPCR and pluripotency of control_2 was evaluated by quantitative PCR analysis of Oct 3/4, Sox2, Klf4, and c-Myc using SYBR green. Cells were passaged every 45days at 1:10 or 1:12 ratio using accutase (Nacalai Tesque, Cat.# 12679-54). Dissociated cells were seeded on Matrigel-coated 6-well plates. The medium was supplemented with 5M Y27632 (TOCRIS, Cat.# 1254), a Rho-associated kinase inhibitor, on the first day of each passage. All cell lines were authenticated by their name, checked their sterrility regularly, and monitored of mycoplasma contamination using by PCR kit (Minerva biolabs, Cat.# 11-9025).

CMs were differentiated from hiPSCs using a previously reported protocol33. Briefly, hiPSCs were seeded into a 12-well growth-factor-reduced (GFR) Matrigel-coated plate, grown for 4days at 37C in 5% CO2 and mTeSR1 medium, and allowed to reach 8090% confluency. On day 0 of differentiation, the medium was changed to differentiation media, which was RPMI containing 2% B27 minus insulin supplement (Gibco, Cat.# A18956-01) and 1012M CHIR99021 (Selleck, Cat.# S2924), a GSK3 inhibitor. After incubation for 24h, the medium was replaced with fresh differentiation medium. On day 3, the medium was replaced with differentiation medium containing 5M IWP-2 (TOCRIS, Cat.# 3533), a Wnt inhibitor. On day 5, the medium was replaced with fresh differentiation medium. On day 7, B27 minus insulin was replaced with a B27 supplement (Gibco, Cat.# 17504044). Differentiated hiPSC-CMs were purified in glucose-depleted lactate medium as described previously34.

C2C12 cells were kindly provided by Dr. Yuji Yamanashi (The Institute of Medical Science, The University of Tokyo)35. The growth medium was DMEM/F12 (Sigma-Aldrich, Cat.# D6421) containing 20% FBS, 2mM glutamine (Gibco, Cat.# 25030081), 100 units/mL penicillin, and 100g/mL streptomycin. Cells were incubated at 37C in a humidified incubator containing 5% CO2. Myoblasts were differentiated into myotubes in DMEM/F12 medium containing 2% horse serum (Gibco, Cat.# 16050122, Lot. 1968945)36,37.

Lenti-CRISPR v2 (Addgene, Cat. # 52961), which contains a puromycin resistance gene, carrying a guide RNA oligonucleotide (5-acctcacccttcagtcacgg-3 for Exon 5 of the Hoil-1l gene or 5-acgcagcaccacggcctcgc-3 for Exon 7 of the Hoil-1l gene) was constructed. HEK293T cells were transfected with the plasmids using Lipofectamine 2000 (Thermo Fisher, Cat.# 11668019). Viruses were harvested at 48h after transfection, and the media were filtered through a 0.45m PES filter. C2C12 cells were transduced with the viruses in medium containing 10g/mL polybrene. At 24h after transduction, puromycin selection was started. The selected cells were collected and KO of Hoil-1l was confirmed by Sanger DNA sequencing.

Myotubes differentiated from C2C12 cells on day 5 of differentiation were fixed in 4% paraformaldehyde (PFA) for 1h at 4C, permeabilized in 0.1% Triton X-100 for 10min at room temperature, and blocked in PBS containing 3% skim milk for 1h. Thereafter, myotubes were stained with an anti-MHC antibody (1:200, mouse monoclonal, R&D Systems, Cat.# MAB4470). The fusion index was calculated by dividing the number of nuclei in myotubes by the total number of nuclei in a field of view36. The MHC density was calculated by dividing the area occupied by MHC-positive myotubes by the total area of the field of view. The fusion index and MHC density were reported as averages of at least three fields of view (>500 total nuclei). Three independent experiments were performed for the calculation. For pluripotency marker analysis, undifferentiated hiPSC colonies were fixed in the same way, and fixed cells were stained with mouse anti-Oct3/4 (1:50, Santa Cruz Biotechnology, Cat.# sc5279) and anti-TRA1-81 (1:100, Millipore, Cat.# MAB4381) antibodies. Cells were then incubated with Alexa Fluor-conjugated secondary antibodies (1:1000) overnight at 4C.Nuclei were stained withHoechst 33342(1:1000, Invitrogen, Cat.#H3570). For immunofluorescence microscopy analysis of hiPSC-CMs, size and multinucleation were analyzed after around 5060days of differentiation and mitosis was analyzed after 20days of differentiation. hiPSC-CMs were replated onto GFR Matrigel-coated 24-well dishes, incubated at 37C in 5% CO2 for 72h, fixed in 4% PFA for 1h at 4C, permeabilized in 0.1% Triton X-100 for 10min at room temperature, and blocked in PBS containing 3% skim milk for 1h. Thereafter, hiPSC-CMs were stained with anti-cTnT (1:100, mouse monoclonal, Thermo Fisher, Cat.# MA5-12960) and anti-phospho-histone H3 (Ser10) (1:1000, rabbit monoclonal, Cell Signaling, Cat.# D7N8E) antibodies. After primary antibody treatment, cells were rinsed three times with PBS for 5min at room temperature and then incubated overnightat 4 with secondary antibodies diluted 1:1000 in PBS. Nuclei were stained with Hoechst 33342. For isotype controls, mouse IgG1 isotype (BD Biosciences, Cat.# 554121) and rabbit IgG isotype (BD Biosciences, Cat.# 550875) were used. All immunofluorescence analyses were performed using a BZ-710X microscope (Keyence). The TUNEL assay was performed using a Cell Death Detection Kit (Roche, Cat.# 11684795910) following the manufacturers protocol.

Myotubes at day 5 of differentiation were lysed in M-PER buffer (Thermo Scientific, Cat.# 78501) containing 1protease inhibitor and then incubated on ice. The samples were sonicated on ice for 30s. The lysates were incubated on ice for 10min and then centrifuged at 15,000rpm for 15min. Protein concentrations were determined using the Bradford assay. Thereafter, 30g of protein was loaded onto each lane of 10% SDS-PAGE gels. The membranes were probed with an anti-MHC antibody (1:100, R&D Systems, Cat.# MAB4470) in blocking buffer (5% BSA) at 4C overnight, washed, incubated in secondary antibodies for 1h at room temperature, developed using ECL western blotting substrate (Bio-Rad, Cat.# 1705060), and imaged using the ChemiDoc MP Imaging System (Bio-Rad). The blots were cut prior to hybridization with antibodies, and two replicates were done at the same time for Fig.1B and Supplementary Fig.2C as shown in supplementaryFig.5.

hiPSC-CMs were dissociated on the day of evaluation by incubating them in 0.25% trypsinEDTA for 1015min at 37C. They were fixed in Cytofix/Cytoperm solution (BD Biosciences, Cat.# 554714) for 20min at 4C, washed with BD Perm/Wash buffer (Cat.# 554723), stained with an anti-cTnT antibody (1:200, mouse monoclonal, Thermo Fisher, Cat.# MA5-12960) followed by Alexa Fluor-conjugated secondary antibodies, and analyzed using FACSverse (BD Biosciences). In cell cycle analysis, hiPSC-CMs after 20days of differentiation were gathered. After fixing and washing the hiPSC-CMs as described above, they were stained with an anti-cTnT antibody (1:200) and an anti-Ki67 antibody (1:400, rabbit monoclonal, Cell Signaling Technology, Cat. # 9129) followed by Alexa Fluor-conjugated secondary antibodies, and analyzed using FACSverse. Data were collected from at least 10,000 events. Data with>70% cTnT populations were used for all experimental analyses.

Total RNA was extracted from day 0 to day 7 of myotube differentiation using an RNeasy Mini Kit (Qiagen, Cat.# 74104) according to the manufacturers instructions. qPCR was performed using SYBR Green PCR Master Mix (Takara, Cat.# RR820) on a StepOnePlus system (Thermo Fisher Scientific) with the Ct method. GAPDH was used to standardize gene expression. Total RNA was extracted from hiPSC-CMs at 4560days after differentiation.

Hoil-1l-KO and control mouse embryos were generated as described previously14. Paraffin sections of E10.5 Hoil-1l null/+ and control littermate mouse embryos were deparaffinized and stained with H&E.

RNA was isolated from C2C12 cells using a RNeasy Mini Kit, according to the manufacturers instructions. RNA integrity was measured using an Agilent 2200 TapeStation and RNA Screen Tapes (Agilent Technologies). Sequencing libraries were prepared using a NEBNext Ultra II RNA Library Kit for Illumina (New England Biolabs) with the NEBNext Poly (A) mRNA Magnetic Isolation Module (New England Biolabs), according to the manufacturers protocol. Prepared libraries were run on an Illumina HiSeq X sequencing platform in 150bp paired-end mode. Sequencing reads were aligned to the GRCm38 mouse genome assembly using STAR (c.2.5.3). Mapped reads were counted for each gene using the GenomonExression pipeline (https://github.com/Genomon-Project/GenomonExpression). Normalization of the read counts of RNA seq data and differential expression analysis were performed using the Bioconductor package DESeq2 (version 1.26.0). Differentially expressed genes with a greater than twofold change and a false discovery rate less than 0.1 were filtered and evaluated. RNA seq data have been deposited with links to BioProject accession number PRJDB17426 in the DDBJ BioProject database.

GSEA was performed using software (version 4.0.3) from the Broad Institute. Normalized expression data obtained from RNA seq were assessed using GSEA software and the Molecular Signature Database (http://www.broad.mit.edu/gsea/). c5 ontology gene sets were used, and a false discovery rate less than 0.01 was considered to be statistically significant. Pathway enrichment analysis using g:Profiler and visualization of enrichment results in an enrichment map were performed using Cytoscape software (version 3.7.2) as described previously38.

Data are shown as meanSEMs, as indicated in the figure legends. All statistical analyses were performed using Welchs t-test with GraphPad Prism (version 9.00, GraphPad Software). P<0.05 was defined as significant.

Use of patient-derived iPSCs was approved by the Ethics Committee of Kyoto University (R0091 and G0687), and written informed consent obtained from the donor (or their guardians) in accordance with the Declaration of Helsinki.

View original post here:
HOIL-1L deficiency induces cell cycle alteration which causes immaturity of skeletal muscle and cardiomyocytes ... - Nature.com

Duke team completes ten-year study on gene expression in stem cells – Duke Chronicle

How do cells acquire their identities? In hopes of answering this question, a Duke team recently completed a study explaining the expression of stem cells after a decade of research.

Stem cells are found in every living organism, including humans and plants. They are initially unspecialized and can develop into almost any cell type while dividing to produce new cells over time. At this point, theyre faced with a choice: divide to make copies of themselves or create something new.

To explore how stem cells make this decision, the group researched the expression of stem cells in plants by developing a specialized microscope that takes precise photographs through a technique called light sheet microscopy.

Cara Winter, associate research professor in biology, and postdoctoral associate Pablo Szekely began working on the project in the lab of Philip Benfey, former Paul Kramer distinguished professor of biology who passed away last year.

This eventually grew to a collaboration with the California Institute of Technology, which included a microscopist, a computer scientist, graduate students and undergraduate student assistants. Winter emphasized that the project was a collaborative effort.

There's no one person that knows all of the information that you need to know to make the microscope and the project work. So you really have to work together to figure out how to get to the goal, Winter said.

Researchers at the California Institute of Technology visited the Duke lab to help construct the microscope. Compared to traditional imaging, this specialized light sheet microscope caused less toxicity and photobleaching, which allowed the team to image cells for longer. This was key because the microscope needed a long observation period to collect enough data on the cells.

The microscope tracked the changing colors of the root cells, which indicated whether the cell would divide and the type of division that would occur. The researchers then connected this data with the level of proteins in the cells to explore how those proteins were associated with cell division. The level of high-resolution imaging of stem cells over such a significant period has never been done before.

Szekely noted that this was made possible by using machine learning and other technological tools to answer these biological questions.

The study allowed the team to photograph the expression of multiple genes in the context of a living organism and connect that gene expression to cell division. They were also able to use that data to test a model of a gene regulatory network, which allowed them to gain insight into asymmetric cell division and how it interfaces with the cell cycle.

The groups results were published in Nature. The study's findings connect to further research for humans and other animals in cancer therapies, drugs, and other aspects of the cell cycle.

I'm hoping to continue following with the next steps of this project, following up on sort of the ideas that came out of the paper, trying to understand exactly what these two regulators are doing early in the cell cycle, Winter said. I'm hoping to stick with imaging, continuing imaging and developing new imaging technologies to ask questions that couldn't be asked.

Signup for our weekly newsletter. Cancel at any time.

Aseel Ibrahim is a Trinity first-year and a staff reporter for the news department.

Read more from the original source:
Duke team completes ten-year study on gene expression in stem cells - Duke Chronicle

SCG CELL THERAPY AND A*STAR LAUNCH JOINT LABS WITH COLLABORATION NEARING S$30 MILLION TO … – PR Newswire

SINGAPORE, April 16, 2024 /PRNewswire/ --SCG Cell Therapy (SCG) and the Agency for Science, Technology and Research (A*STAR) announced the launch joint laboratories for cellular immunotherapies. This collaboration, at a combined funding of close to S$30 million supported under Singapore's Research, Innovation and Enterprise 2025 Plan (RIE2025), aims to advance the development of induced pluripotent stem cell (iPSC) technology to produce novel cell therapies that meet Good Manufacturing Practice (GMP) standards. The collaboration will also establish a talent development programme to train the next generation of experts in this field, in accordance with current GMP and regulatory requirements.

The research and application of new technologies are essential for addressing growing healthcare needs and maintaining long-term sustainability. However, turning laboratory innovations into practical clinical solutions poses significant challenges. These often involve developing manufacturing processes, validating analytical methods, and implementing automation and digitalisation to guarantee the stability and scalability of products.

The joint laboratories, established at SCG's GMP facility and A*STAR's research facility, leverage SCG's and A*STAR's proprietary technologies to develop scalable GMP-grade iPSC and therapeutic products. SCG contributes its specialised, automated cell therapy manufacturing technologies, while A*STAR brings its unique monoclonal antibody assets, iPSC banks, and expertise in process scaling and analytics.

This collaboration bridges the expertise between public sector research and development (R&D) and industry, consolidating resources from SCG Cell Therapy and A*STAR's Bioprocessing Technology Institute (BTI) and Institute of Molecular and Cell Biology (IMCB) to advance innovative R&D towards GMP manufacturing. Additionally, it immerses researchers in the rigorously controlled GMP environment, facilitating the progression from research to clinical application.

"Cellular immunotherapies herald a new era of regenerative medicine, offering hope for patients with cancers and other serious illnesses. As a key player in T cell receptor (TCR) T cell therapeutics, SCG has developed in-house cGMP manufacturing capabilities to supply high-quality cell therapy products to patients. Through this first-of-its-kind joint collaboration with A*STAR, we bring together A*STAR's advanced iPSC technology and bioprocessing capabilities with our expertise in GMP cell therapy manufacturing and clinical development, furthering our mission to provide affordable off-the-shelf cell therapy treatment options to patients", said Christy Ma, Chief Strategy Officer of SCG Cell Therapy.

"The discovery of iPSCs has revolutionised regenerative medicine, offering the potential for standardised, off-the-shelf cell therapies. Through this collaboration with SCG Cell Therapy, we aim to accelerate the translation of iPSC research into clinically viable therapies and strengthen Singapore's position as a global leader in cell therapy innovation. By leveraging our complementary expertise and resources, the joint labs will not only advance iPSC technology for scalable, GMP-compliant cell therapy production but also serve as a platform for nurturing the next generation of talent in this transformative field," said Prof Koh Boon Tong, Executive Director, A*STAR's BTI.

About iPSC

Induced pluripotent stem cells (also known as iPS cells or iPSCs) are a type of pluripotent stem cell that can be generated directly from adult cells. The iPSC technology was pioneered by Shinya Yamanaka's lab in Kyoto, Japan, who showed in 2006 that the introduction of four specific genes encoding transcription factors could convert adult cells into pluripotent stem cells. He was awarded the 2012 Nobel Prize along with Sir John Gurdon "for the discovery that mature cells can be reprogrammed to become pluripotent". Pluripotent stem cells hold great promise in the field of regenerative medicine. Because they can propagate indefinitely, as well as give rise to every other cell type in the body (such as neurons, heart, pancreatic and liver cells), they represent a single source of cells that could be used to replace those lost to damage or disease.

About SCG Cell Therapy

SCG is a clinical-stage biotechnology company focusing on the development of novel immunotherapies in infections and its associated cancers. The company targets the most common cancer-causing infections: helicobacter pylori, human papillomavirus, and hepatitis B, and develops a broad and unique pipeline against infections and to prevent and cure its associated cancers. Established and headquartered in Singapore, SCG combines regional advantages in Singapore, China and Germany, covering the entire value chain from innovative drug research and discovery, manufacturing, clinical development and commercialization. For more information about SCG, please visit us at http://www.scgcell.com.

About the Agency for Science, Technology and Research (A*STAR)

The Agency for Science, Technology and Research (A*STAR) is Singapore's lead public sector R&D agency. Through open innovation, we collaborate with our partners in both the public and private sectors to benefit the economy and society. As a Science and Technology Organisation, A*STAR bridges the gap between academia and industry. Our research creates economic growth and jobs for Singapore, and enhances lives by improving societal outcomes in healthcare, urban living, and sustainability. A*STAR plays a key role in nurturing scientific talent and leaders for the wider research community and industry. A*STAR's R&D activities span biomedical sciences to physical sciences and engineering, with research entities primarily located in Biopolis and Fusionopolis. For ongoing news, visit http://www.a-star.edu.sg.

Follow us on

Facebook| LinkedIn| Instagram| YouTube|TikTok

SOURCE SCG Cell Therapy Pte Ltd

Original post:
SCG CELL THERAPY AND A*STAR LAUNCH JOINT LABS WITH COLLABORATION NEARING S$30 MILLION TO ... - PR Newswire