Category Archives: Induced Pluripotent Stem Cells

Imaging cAMP nanodomains in human iPSC-derived cardiomyocytes – Nature.com

Cardiac activity is regulated by the -adrenergic pathway. The activation of this pathway triggers a cellular signalling cascade that increases the production of cAMP, a cyclic nucleotide that activates the enzyme protein kinase A (PKA). PKA phosphorylates key proteins involved in cellular contraction, but can also phosphorylate a multitude of other proteins with different functions. To achieve specific effects, cAMP is confined in nanoscale subcellular domains (nanodomains) close to PKA and its targets. The maintenance and regulation of these nanodomains are central to functional signal transduction, and their dysregulation can result in diseases such as heart failure.

I use this technique in human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to study how the maturation of these cells is affected by a newly identified cAMP nanodomain found at gap junctions, which regulate the communication between adjacent cardiomyocytes. To understand the role of the gap junction-associated cAMP nanodomain in human iPSC-derived cardiomyocytes, endogenous levels of protein expression must be maintained to avoid interference with their maturation process. This technique can more broadly be used to study cAMP nanodomains in which overexpression of the target protein might impair cell physiology. This tool will provide unique insights into the processes involved in human iPSC-derived cardiomyocyte maturation and can also be used to identify new targets in the -adrenergic pathway that might be relevant for the treatment of diseases, such as heart failure.

Originally posted here:
Imaging cAMP nanodomains in human iPSC-derived cardiomyocytes - Nature.com

Colossal Creates Elephant Stem Cells for the First Time in Quest to Revive the Woolly Mammoth – Singularity Hub

The last woolly mammoth roamed the vast arctic tundra 4,000 years ago. Their genes still live on in a majestic animal todaythe Asian elephant.

With 99.6 percent similarity in their genetic makeup, Asian elephants are the perfect starting point for a bold plan to bring the mammothor something close to itback from extinction. The project, launched by biotechnology company Colossal in 2021, raised eyebrows for its moonshot goal.

The overall playbook sounds straightforward.

The first step is to sequence and compare the genomes of mammoth and elephant. Next, scientists will identify the genes behind the physical traitslong hair, fatty depositsthat allowed mammoths to thrive in freezing temperatures and then insert them into elephant cells using gene editing. Finally, the team will transfer the nucleuswhich houses DNAfrom the edited cells into an elephant egg and implant the embryo into a surrogate.

The problem? Asian elephants are endangered, and their cellsespecially eggsare hard to come by.

Last week, the company reported a major workaround. For the first time, they transformed elephant skin cells into stem cells, each with the potential to become any cell or tissue in the body.

The advance makes it easier to validate gene editing results in the lab before committing to a potential pregnancywhich lasts up to 22 months for elephants. Scientists could, for example, coax the engineered elephant stem cells to become hair cells and test for gene edits that give the mammoth its iconic thick, warm coat.

These induced pluripotent stem cells, or iPSCs, have been especially hard to make from elephant cells. The animals are a very special species and we have only just begun to scratch the surface of their fundamental biology, said Dr. Eriona Hysolli, who heads up biosciences at Colossal, in a press release.

Because the approach only needs a skin sample from an Asian elephant, it goes a long way to protecting the endangered species. The technology could also support conservation for living elephants by providing breeding programs with artificial eggs made from skin cells.

Elephants might get the hardest to reprogram prize, said Dr. George Church, a Harvard geneticist and Colossal cofounder, but learning how to do it anyway will help many other studies, especially on endangered species.

Nearly two decades ago, Japanese biologist Dr. Shinya Yamanaka revolutionized biology by restoring mature cells to a stem cell-like state.

First demonstrated in mice, the Nobel Prize-winning technique requires only four proteins, together called the Yamanaka factors. The reprogrammed cells, often derived from skin cells, can develop into a range of tissues with further chemical guidance.

Induced pluripotent stem cells (iPSCs), as theyre called, have transformed biology. Theyre critical to the process of building brain organoidsminiature balls of neurons that spark with activityand can be coaxed into egg cells or models of early human embryos.

The technology is well-established for mice and humans. Not so for elephants. In the past, a multitude of attempts to generate elephant iPSCs have not been fruitful, said Hysolli.

Most elephant cells died when treated with the standard recipe. Others turned into zombie senescent cellsliving but unable to perform their usual biological functionsor had little change from their original identity.

Further sleuthing found the culprit: A protein called TP53. Known for its ability to fight off cancer, the protein is often dubbed the genetic gatekeeper. When the gene for TP53 is turned on, the protein urges pre-cancerous cells to self-destruct without harming their neighbors.

Unfortunately, TP53 also hinders iPSC reprogramming. Some of the Yamanaka factors mimic the first stages of cancer growth which could cause edited cells to self-destruct. Elephants have a hefty 29 copies of the protector gene. Together, they could easily squash cells with mutated DNA, including those that have had their genes edited.

We knew p53 was going to be a big deal, Church told the New York Times.

To get around the gatekeeper, the team devised a chemical cocktail to inhibit TP53 production. With a subsequent dose of the reprogramming factors, they were able to make the first elephant iPSCs out of skin cells.

A series of tests showed the transformed cells looked and behaved as expected. They had genes and protein markers often seen in stem cells. When allowed to further develop into a cluster of cells, they formed a three-layered structure critical for early embryo development.

Weve been really waiting for these things desperately, Church told Nature. The team published their results, which have not yet been peer-reviewed, on the preprint server bioRxiv.

The companys current playbook for bringing back the mammoth relies on cloning technologies, not iPSCs.

But the cells are valuable as proxies for elephant egg cells or even embryos, allowing the scientists to continue their work without harming endangered animals.

They may, for example, transform the new stem cells into egg or sperm cellsa feat so far only achieved in micefor further genetic editing. Another idea is to directly transform them into embryo-like structures equipped with mammoth genes.

The company is also looking into developing artificial wombs to help nurture any edited embryos and potentially bring them to term. In 2017, an artificial womb gave birth to a healthy lamb, and artificial wombs are now moving towards human trials. These systems would lessen the need for elephant surrogates and avoid putting their natural reproductive cycles at risk.

As the study is a preprint, its results havent yet been vetted by other experts in the field. Many questions remain. For example, do the reprogrammed cells maintain their stem cell status? Can they be transformed into multiple tissue types on demand?

Reviving the mammoth is Colossals ultimate goal. But Dr. Vincent Lynch at the University of Buffalo, who has long tried to make iPSCs from elephants, thinks the results could have a broader reach.

Elephants are remarkably resistant to cancer. No one knows why. Because the studys iPSCs are stripped of TP53, a cancer-protective gene, they could help scientists identify the genetic code that allows elephants to fight tumors and potentially inspire new treatments for us as well.

Next, the team hopes to recreate mammoth traitssuch as long hair and fatty depositsin cell and animal models made from gene-edited elephant cells. If all goes well, theyll employ a technique like the one used to clone Dolly the sheep to birth the first calves.

Whether these animals can be called mammoths is still up for debate. Their genome wont exactly match the extinct species. Further, animal biology and behavior strongly depend on interactions with the environment. Our climate has changed dramatically since mammoths went extinct 4,000 years ago. The Arctic tundratheir old homeis rapidly melting. Can the resurrected animals adjust to an environment they werent adapted to roam?

Animals also learn from each other. Without a living mammoth to show a calf how to be a mammoth in its natural habitat, it may adopt a completely different set of behaviors.

Colossal has a general plan to tackle these difficult questions. In the meantime, the work will help the project make headway without putting elephants at risk, according to Church.

This is a momentous step, said Ben Lamm, cofounder and CEO of Colossal. Each step brings us closer to our long-term goals of bringing back this iconic species.

Image Credit: Colossal Biosciences

Go here to see the original:
Colossal Creates Elephant Stem Cells for the First Time in Quest to Revive the Woolly Mammoth - Singularity Hub

How stem cells might be used in planned de-extinction of woolly mammoth – Cosmos

Sometimes it takes the smallest thing to undertake a mammoth task.

Thats what researchers behind the attempts to de-extinct the woolly mammoth are hoping as they announced what they believe to be a step forward in their efforts.

One Texas-based company has made de-extinction its business. It is looking at not only de-extinction of the woolly mammoth, but also dodos and Australias thylacine both hunted to extinction within the last 500 years.

The key is an announcement this week from researchers with Colossal Biosciences who say theyve derived induced pluripotent stem cells (iPSCs) from Asian elephants (Elephas maximus).

These iPSCs are reprogrammed to be able to give rise to any cell type in the body.

It means the researchers might be able to investigate the genetic differences between the woolly mammoth (Mammathus primigenius) and their closest living relatives the Asian elephant. They can also test gene edits without needing tissue from living animals.

Woolly mammoths roamed Earth for nearly 800,000 years.

They diversified from the steppe mammoth (Mammuthus trogontherii) at the beginning of the Middle Pleistocene (770,000126,000 years ago). They were closely related to the North American Columbian mammoth (Mammathus columbi) and DNA studies show they occasionally interbred.

Woolly mammoths were up to 3.5 metres tall at the shoulder and could weigh as much as 8 metric tons. (By contrast the Asian elephant is 2-3m and weights up to 5t.)

Mammoths are synonymous with the last Ice Age which ended about 12,000 years ago. Its believed that a combination of a warming climate and human hunting saw woolly mammoth numbers decline.

They died out so recently that some mammoth bodies have been recovered extremely well preserved in ice and snow.

The last stronghold of the woolly mammoth was the Siberian island of Wrangel where they lived until as recently as 4,000 years ago.

When these last mammoths died, the Great Pyramids of Giza were already 600 years old. Stonehenge had been around for 1,000 years and Sumerian poets had begun compiling the works that would over the next 800 years be brought together into the Epic of Gilgamesh.

In the great scheme of geological time, we are tantalisingly close to these remarkable creatures.

The successful formation of Asian elephant iPSCs in the lab is critical to understanding how the woolly mammoths genetic code sets it apart from its modern counterparts.

Which bits of DNA come together to produce features like their shaggy hair, curved tusks, fat deposits and dome skulls? These are the kinds of questions scientists at Colossal now feel they can answer.

It is also possible that the iPSCs can lead to producing elephant sperm and egg cells in the lab. Anyone whos had the birds and the bees chat doesnt need to be told why thats important in de-extinction.

Being able to create these cells in a lab is particularly important given the precariousness of Asian elephant populations.

Fewer than 50,000 Asian elephants remain in the wild according to the World Wildlife Fund. They are listed as endangered on the IUCNs Red List. Attempts to retrieve egg and sperm cells from Asian elephants would be difficult and potentially adverse.

Making elephant iPSCs has been so difficult because of complex gene pathways unique to these animals. Colossals genetic engineers overcame this by suppressing core genes called TP53 which regulate cell growth and halt the duplicating process.

But the work doesnt stop.

Theyre still looking at alternative methods to create iPSCs and maturing the ones theyve already made.

Theres also a lot still to learn about the complex 22-month gestation period of elephants if a healthy woolly mammoth calf is to be produced through in vitro fertilisation of a modern elephant.

Colossals plan is to have a living, breathing woolly mammoth by 2028.

For that to happen, the company is also looking into restoring suitable tundra steppe habitats in Canada and the US where the reborn mammoth population can settle.

Read the original:
How stem cells might be used in planned de-extinction of woolly mammoth - Cosmos

The long and winding road of reprogramming-induced rejuvenation – Nature.com

Vos, T. et al. Global Burden of 369 Diseases and Injuries in 204 Countries and territories, 19902019: a Systematic Analysis for the Global Burden of Disease Study 2019. Lancet 396, 12041222 (2020).

Article Google Scholar

Yang, J. H. et al. Loss of epigenetic information as a cause of mammalian aging. Cell 186, 305326 (2023).

Article CAS PubMed PubMed Central Google Scholar

Duffield T. et al Epigenetic fidelity in complex biological systems and implications for ageing. Biorxiv. Published online April 30 https://doi.org/10.1101/2023.04.29.538716 (2023).

Kerepesi, C., Zhang, B., Lee, S. G., Trapp, A. & Gladyshev, V. N. Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Sci. Adv. 7, eabg6082 (2021).

Article ADS CAS PubMed PubMed Central Google Scholar

Zhang B. et al. Epigenetic profiling and incidence of disrupted development point to gastrulation as aging ground zero in Xenopus laevis. Biorxiv. Published online August 4 https://doi.org/10.1101/2022.08.02.502559 (2022).

Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013).

Article PubMed PubMed Central Google Scholar

Simpson D. J., Olova N. N., Chandra T. Cellular reprogramming and epigenetic rejuvenation. Clin. Epigenetics. 13, https://doi.org/10.1186/s13148-021-01158-7 (2021).

Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 17191733.e12 (2016).

Article CAS PubMed PubMed Central Google Scholar

Manukyan M., Singh P. B. Epigenome rejuvenation: HP1 mobility as a measure of pluripotent and senescent chromatin ground states. Sci. Rep. 4, https://doi.org/10.1038/srep04789 (2014).

Sarkar, T. J. et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat. Commun. 11, 1545 (2020).

Article ADS CAS PubMed PubMed Central Google Scholar

Chondronasiou, D. et al. Multiomic rejuvenation of naturally aged tissues by a single cycle of transient reprogramming. Aging Cell 21, e13578 (2022).

Article CAS PubMed PubMed Central Google Scholar

Gill, D. et al. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. eLife 11, e71624 (2022).

Article CAS PubMed PubMed Central Google Scholar

Olova, N., Simpson, D. J., Marioni, R. E. & Chandra, T. Partial reprogramming induces a steady decline in epigenetic age before loss of somatic identity. Aging Cell 18, e12877 (2018).

Article PubMed PubMed Central Google Scholar

Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124129 (2020).

Article ADS CAS PubMed PubMed Central Google Scholar

Browder, K. C. et al. In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat. Aging 2, 243253 (2022).

Article CAS PubMed Google Scholar

Macip C. C. et al. Gene therapy mediated partial reprogramming extends lifespan and reverses age-related changes in aged mice. Biorxiv. Published online January 5, 2023. https://doi.org/10.1101/2023.01.04.522507.

Pupo, A. et al. AAV vectors: The Rubiks cube of human gene therapy. Mol. Ther.: J. Am. Soc. Gene Ther. 30, 35153541 (2022).

Article CAS Google Scholar

Wang, C. et al. In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche. Nat. Commun. 12, 3094 (2021).

Article ADS CAS PubMed PubMed Central Google Scholar

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

Article ADS CAS PubMed Google Scholar

Schoenfeldt L. et al. Chemical reprogramming ameliorates cellular hallmarks of aging and extends lifespan. Biorxiv. Published online August 31, (2022). https://doi.org/10.1101/2022.08.29.505222.

Guan J. et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature 605, 325331 (2022).

Liuyang, Shijia et al. Highly efficient and rapid generation of human pluripotent stem cells by chemical reprogramming. Cell Stem Cell 30, 450459.e9 (2023).

Article CAS PubMed Google Scholar

Mitchell W. et al. Multi-omics characterization of partial chemical reprogramming reveals evidence of cell rejuvenation. Biorxiv. Published online June 30, 2023. https://doi.org/10.1101/2023.06.30.546730.

Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53p21 pathway. Nature 460, 11321135 (2009).

Article ADS CAS PubMed PubMed Central Google Scholar

Levine, A. J., Puzio-Kuter, A. M., Chan, C. S. & Hainaut, P. The role of the p53 protein in stem-cell biology and epigenetic regulation. Cold Spring Harb. Perspect. Med. 6, a026153 (2016).

Article PubMed PubMed Central Google Scholar

Tyner, S. D. et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 415, 4553 (2002).

Article ADS CAS PubMed Google Scholar

Hu, X., Eastman, A. E. & Guo, S. Cell cycle dynamics in the reprogramming of cellular identity. FEBS Lett. 593, 28402852 (2019).

Article CAS PubMed Google Scholar

Mosteiro, Lluc, Pantoja, C., de Martino, Alba & Serrano, M. Senescence promotes in vivo reprogramming through p16INK 4a and IL-6. Aging Cell 17, e12711e12711 (2017).

Article PubMed PubMed Central Google Scholar

Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Sci. (N. Y., NY) 354, aaf4445 (2016).

Article Google Scholar

Chen, Y. et al. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science 373, 15371540 (2021).

Article ADS CAS PubMed Google Scholar

Lpez-Otn, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: An expanding universe. Cell 186, 243278 (2023).

Article PubMed Google Scholar

Horvath, S. et al. Epigenetic clock for skin and blood cells applied to Hutchinson Gilford Progeria Syndrome and ex vivo studies. Aging 10, 17581775 (2018).

Article CAS PubMed PubMed Central Google Scholar

Kabacik, S. et al. The relationship between epigenetic age and the hallmarks of aging in human cells. Nat. Aging 2, 484493 (2022).

Article PubMed PubMed Central 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

Rouhani, F. J. et al. Substantial somatic genomic variation and selection for BCOR mutations in human induced pluripotent stem cells. Nat. Genet. 54, 14061416 (2022).

Article CAS PubMed PubMed Central Google Scholar

DAntonio, M. et al. Insights into the mutational burden of human induced pluripotent stem cells from an integrative multi-omics approach. Cell Rep. 24, 883894 (2018).

Article PubMed PubMed Central Google Scholar

Kosanke, M. et al. Reprogramming enriches for somatic cell clones with small-scale mutations in cancer-associated genes. Mol. Ther.: J. Am. Soc. Gene Ther. 29, 25352553 (2021).

Article CAS Google Scholar

Young, MargaretA. et al. Background mutations in parental cells account for most of the genetic heterogeneity of induced pluripotent stem cells. Cell Stem Cell 10, 570582 (2012).

Article CAS PubMed PubMed Central Google Scholar

Poganik J. R. et al. Biological age is increased by stress and restored upon recovery. Cell Metab. Published online April 4, 2023:S1550-4131(23)000931. https://doi.org/10.1016/j.cmet.2023.03.015.

Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 49, 359367 (2013).

Article CAS PubMed Google Scholar

Levine, M. E. et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY) 10, 573591 (2018).

Article PubMed Google Scholar

Lu, A. T. et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11, 303327 (2019).

Article CAS PubMed PubMed Central Google Scholar

Ying K. et al. Causality-enriched epigenetic age uncouples damage and adaptation. Nature Aging. Published online January 19, 2024. https://doi.org/10.1038/s43587-023-00557-0.

Nikopoulou, C. et al. Spatial and single-cell profiling of the metabolome, transcriptome and epigenome of the aging mouse liver. Nat. Aging 3, 14301445 (2023).

Article CAS PubMed PubMed Central Google Scholar

Marin, R. M. et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 460, 11491153 (2009).

Article ADS PubMed PubMed Central Google Scholar

Liu, X. et al. Yamanaka factors critically regulate the developmental signaling network in mouse embryonic stem cells. Cell Res. 18, 11771189 (2008).

Article CAS PubMed Google Scholar

Arabac, D. H., Terziolu, G., Bayrba, B. & nder, T. T. Going up the hill: Chromatinbased barriers to epigenetic reprogramming. FEBS J. 288, 47984811 (2020).

Article PubMed Google Scholar

Mertens, J., Reid, D., Lau, S., Kim, Y. & Gage, F. H. Aging in a dish: iPSC-derived and directly induced neurons for studying brain aging and age-related neurodegenerative diseases. Annu. Rev. Genet. 52, 271293 (2018).

Article CAS PubMed PubMed Central Google Scholar

Sheng C. et al. A stably self-renewing adult blood-derived induced neural stem cell exhibiting patternability and epigenetic rejuvenation. Nat. Commun. 9, https://doi.org/10.1038/s41467-018-06398-5 (2018).

Shi, G. & Jin, Y. Role of Oct4 in maintaining and regaining stem cell pluripotency. Stem Cell Res. Ther. 1, 39 (2010).

Article CAS PubMed PubMed Central Google Scholar

Kriukov D., Khrameeva E. E., Gladyshev V. N., Dmitriev S. E., Tyshkovskiy A. Longevity and rejuvenation effects of cell reprogramming are decoupled from loss of somatic identity. Biorxiv. Published online December 14, 2022. https://doi.org/10.1101/2022.12.12.520058.

Teshigawara, R., Cho, J., Kameda, M. & Tada, T. Mechanism of human somatic reprogramming to iPS cell. Lab. Investig. 97, 11521157 (2017).

Article CAS PubMed Google Scholar

Wang, B. et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Jdp2-Jhdm1b-Mkk6-Glis1-Nanog-Essrb-Sall4. Cell Rep. 27, 34733485.e5 (2019).

Article CAS PubMed Google Scholar

Adachi, K. et al. Esrrb unlocks silenced enhancers for reprogramming to naive pluripotency. Cell Stem Cell 23, 266275.e6 (2018).

Article CAS PubMed Google Scholar

Carbognin E. et al. Esrrb guides naive pluripotent cells through the formative transcriptional programme. Nat. Cell Biol. 25, 643657 (2023).

Nakagawa, M., Takizawa, N., Narita, M., Ichisaka, T. & Yamanaka, S. Promotion of direct reprogramming by transformation-deficient Myc. Proc. Natl. Acad. Sci. 107, 1415214157 (2010).

Article ADS CAS PubMed PubMed Central Google Scholar

Onder, T. T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598602 (2012).

Continued here:
The long and winding road of reprogramming-induced rejuvenation - Nature.com

Looking for the Path to Safe Cell Rejuvenation – Lifespan.io News

In Nature Communications, Ali Ycel and Vadim Gladyshev have published a review of the current state of the art in partial cellular reprogramming, detailing what this technology does and how it might be used safely.

This paper begins by treading familiar ground on the subject, explaining its end goals and purpose. When successful, partial cellular reprogramming induces reprogramming-induced rejuvenation (RIR), a state in which a cell is transformed into an epigenetically younger cell of the same type and fulfilling the same function [1]. This process has had multiple crucial successes in experimental models, including human muscle cells [2] and skin cells [3] along with restoring vision [4] and extending lifespan [5] in mice.

Much of this work has been done in mice that have been genetically modified to express the necessary factors when doxycycline is administered. This has even been accomplished after birth via an adeno-associated virus (AAV) [5]. While there are four Yamanaka factors, OSKM, the fourth, c-Myc, is often omitted because it raises the risk of cancer. OSK administration significantly reduced the frailty of the treated mice.

As the authors note, applying these sorts of genetic modification techniques directly to human beings is currently infeasible with existing technologies. Partial reprogramming requires carefully determined generation of Yamanaka factors inside cells. To apply this in a clinical setting would require gene therapy that has specific and strong effects on individual tissues, and using the AAV system that works on mice is not yet practical for people [6]. Generating partially reprogrammed cells outside the body, similarly to how induced pluripotent stem cells (iPSCs) are generated, may be feasible for therapeutic purposes.

Administering small molecules to people in order to effect rejuvenation in the form of a drug has been the dream of aging researchers for some time. Previous work has spurred the creation of iPSCs through such chemical means [7]. The authors of this review describe these methods as less powerful than gene therapy and requiring multiple stages of administration. This implies a degree of safety and control that makes them more attractive for human research.

An experiment on mouse cells, which also included Vadim Gladyshev, had revealed that using a 7c cocktail reduced multiple aspects of aging, including epigenetic clock measurements, age-related metabolic changes, and oxidative stress markers [8]. However, it also upregulates the senescence-associated p53 pathway, which is downregulated through normal reprogramming methods and may cause cells to become senescent earlier [9].

Normally, constant expression of the Yamanaka factors in a living organism causes its cells to completely revert to a pluripotent state, in which they forget their roles, become cancerous, and cause the organism to die. For example, inducing OSKM for six days in the hearts of mice was found to be beneficial for them, while extending it for a dozen days proved lethal [10]. However, constantly inducing OSK in neural ganglion cells for a full 10-18 months improved vision without this side effect [4].

The authors note many of the aspects of aging that are improved or possibly improved with RIR, of which the most obvious, epigenetic alterations, is only one. Inflammation and proteostasis are also affected. Telomere attrition, however, occurs only in later reprogramming and is not affected by the partial variety [11]. Direct changes to cellular communication and genomic stability are not yet known.

However, the authors point out that, while full reprogramming does not cells to mutate, creating colonies of iPSCs causes evolutionary pressure: cells with mutations that may not be beneficial for the whole organism may be more prevalent in iPSC colonies [12]. It remains to be seen if this is a concern for partial reprogramming.

The authors also mention a biochemical pluripotency network and the fundamental differences between full and partial rejuvenation. Most critically, they hold that partial reprogramming is caused by factors that are downstream of full reprogramming. If it is possible to directly affect these factors instead of relying on the Yamanaka full-reprogramming factors, it might be possible to cause RIR without risking the dangerous side effects associated with complete reprogramming. However, this area of research remains unexplored.

To do this, we need your support. Your charitable contribution tranforms into rejuvenation research, news, shows, and more. Will you help?

[1] Ocampo, A., Reddy, P., Martinez-Redondo, P., Platero-Luengo, A., Hatanaka, F., Hishida, T., & Belmonte, J. C. I. (2016). In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell, 167(7), 1719-1733.

[2] Sarkar, T. J., Quarta, M., Mukherjee, S., Colville, A., Paine, P., Doan, L., & Sebastiano, V. (2020). Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nature communications, 11(1), 1545.

[3] Gill, D., Parry, A., Santos, F., Okkenhaug, H., Todd, C. D., Hernando-Herraez, I., & Reik, W. (2022). Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. Elife, 11, e71624.

[4] Lu, Y., Brommer, B., Tian, X., Krishnan, A., Meer, M., Wang, C., & Sinclair, D. A. (2020). Reprogramming to recover youthful epigenetic information and restore vision. Nature, 588(7836), 124-129.

[5] Macip, C. C., Hasan, R., Hoznek, V., Kim, J., Lu, Y. R., Metzger IV, L. E., & Davidsohn, N. (2024). Gene Therapy-Mediated Partial Reprogramming Extends Lifespan and Reverses Age-Related Changes in Aged Mice. Cellular Reprogramming, 26(1), 24-32.

[6] Pupo, A., Fernndez, A., Low, S. H., Franois, A., Surez-Amarn, L., & Samulski, R. J. (2022). AAV vectors: The Rubiks cube of human gene therapy. Molecular Therapy.

[7] Guan, J., Wang, G., Wang, J., Zhang, Z., Fu, Y., Cheng, L., & Deng, H. (2022). Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature, 605(7909), 325-331.

[8] Mitchell, W., Goeminne, L. J., Tyshkovskiy, A., Zhang, S., Chen, J. Y., Paulo, J. A., & Gladyshev, V. N. (2023). Multi-omics characterization of partial chemical reprogramming reveals evidence of cell rejuvenation. bioRxiv, 2023-06.

[9] Tyner, S. D., Venkatachalam, S., Choi, J., Jones, S., Ghebranious, N., Igelmann, H., & Donehower, L. A. (2002). p53 mutant mice that display early ageing-associated phenotypes. Nature, 415(6867), 45-53.

[10] Chen, Y., Lttmann, F. F., Schoger, E., Schler, H. R., Zelarayn, L. C., Kim, K. P., & Braun, T. (2021). Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science, 373(6562), 1537-1540.

[11] Takahashi, K., & Yamanaka, S. (2006). Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. cell, 126(4), 663-676.

[12] Kosanke, M., Osetek, K., Haase, A., Wiehlmann, L., Davenport, C., Schwarzer, A., & Martin, U. (2021). Reprogramming enriches for somatic cell clones with small-scale mutations in cancer-associated genes. Molecular Therapy, 29(8), 2535-2553.

Read the original post:
Looking for the Path to Safe Cell Rejuvenation - Lifespan.io News

Lessons from inducible pluripotent stem cell models on neuronal senescence in aging and neurodegeneration – Nature.com

Lpez-Otn, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: an expanding universe. Cell 186, 243278 (2023).

Article PubMed Google Scholar

Hayflick, L. & Moorhead, P. S. The serial cultivation of human diploid cell strains. Exp. Cell. Res. 25, 585621 (1961).

Article CAS PubMed Google Scholar

Harley, C. B., Vaziri, H., Counter, C. M. & Allsopp, R. C. The telomere hypothesis of cellular aging. Exp. Gerontol. 27, 375382 (1992).

Article CAS PubMed Google Scholar

Gorgoulis, V. et al. Cellular senescence: defining a path forward. Cell 179, 813827 (2019).

Article CAS PubMed Google Scholar

Copp, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99118 (2010).

Article PubMed PubMed Central Google Scholar

Herdy, J. R. et al. Increased post-mitotic senescence in aged human neurons is a pathological feature of Alzheimers disease. Cell Stem Cell 29, 16371652 (2022).

Article Google Scholar

Ng, P. Y., McNeely, T. L. & Baker, D. J. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J. 290, 13261339 (2023).

Article CAS PubMed Google Scholar

van Deursen, J. M. The role of senescent cells in ageing. Nature 509, 439446 (2014).

Article ADS PubMed PubMed Central Google Scholar

Crouch, J., Shvedova, M., Thanapaul, R., Botchkarev, V. & Roh, D. Epigenetic regulation of cellular senescence. Cells 11, 672 (2022).

Article CAS PubMed PubMed Central Google Scholar

Kowald, A., Passos, J. F. & Kirkwood, T. B. L. On the evolution of cellular senescence. Aging Cell 19, e13270 (2020).

Article CAS PubMed PubMed Central Google Scholar

Miwa, S., Kashyap, S., Chini, E. & von Zglinicki, T. Mitochondrial dysfunction in cell senescence and aging. J. Clin. Invest. 132, e158447 (2022).

Article CAS PubMed PubMed Central Google Scholar

Muoz-Espn, D. et al. Programmed cell senescence during mammalian embryonic development. Cell 155, 11041118 (2013).

Article PubMed Google Scholar

Gibaja, A. et al. TGF2-induced senescence during early inner ear development. Sci. Rep. 9, 5912 (2019).

Article ADS PubMed PubMed Central Google Scholar

Reichel, W., Hollander, J., Clark, J. H. & Strehler, B. L. Lipofuscin pigment accumulation as a function of age and distribution in rodent brain. J. Gerontol. 23, 7178 (1968).

Article CAS PubMed Google Scholar

Moreno-Garca, A., Kun, A., Calero, O., Medina, M. & Calero, M. An overview of the role of lipofuscin in age-related neurodegeneration. Front. Neurosci. 12, 464 (2018).

Article PubMed PubMed Central Google Scholar

Xu, P. et al. The landscape of human tissue and cell type specific expression and co-regulation of senescence genes. Mol. Neurodegener. 17, 5 (2022).

Article CAS PubMed PubMed Central Google Scholar

Saul, D. et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat. Commun. 13, 4827 (2022).

Article ADS CAS PubMed PubMed Central Google Scholar

Ogrodnik, M. et al. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 20, e13296 (2021).

Article CAS PubMed PubMed Central Google Scholar

Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578582 (2018).

Article ADS CAS PubMed PubMed Central Google Scholar

Musi, N. et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17, e12840 (2018).

Article PubMed PubMed Central Google Scholar

Rocha, L. R. et al. Early removal of senescent cells protects retinal ganglion cells loss in experimental ocular hypertension. Aging Cell 19, e13089 (2020).

Article CAS PubMed Google Scholar

Zhang, P. et al. Senolytic therapy alleviates A-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimers disease model. Nat. Neurosci. 22, 719728 (2019).

Article CAS PubMed PubMed Central Google Scholar

Chinta, S. J. et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinsons disease. Cell Rep. 22, 930940 (2018).

Article CAS PubMed PubMed Central Google Scholar

Gonzales, M. M. et al. Senolytic therapy to modulate the progression of Alzheimers disease (SToMP-AD): a pilot clinical trial. J. Prev. Alzheimers Dis. 9, 2229 (2022).

CAS PubMed Google Scholar

Dehkordi, S. K. et al. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat. Aging 1, 11071116 (2021).

Article PubMed PubMed Central Google Scholar

Langfelder, P. & Horvath, S. Eigengene networks for studying the relationships between co-expression modules. BMC Syst. Biol. 1, 54 (2007).

Article PubMed PubMed Central Google Scholar

Geng, Y. Q., Guan, J. T., Xu, X. H. & Fu, Y. C. Senescence-associated beta-galactosidase activity expression in aging hippocampal neurons. Biochem. Biophys. Res. Commun. 396, 866869 (2010).

Article CAS PubMed Google Scholar

Bhanu, M. U., Mandraju, R. K., Bhaskar, C. & Kondapi, A. K. Cultured cerebellar granule neurons as an in vitro aging model: topoisomerase II as an additional biomarker in DNA repair and aging. Toxicol. In Vitro 24, 19351945 (2010).

Article PubMed Google Scholar

de Mera-Rodrguez, J. A. et al. Endogenous pH 6.0 -galactosidase activity is linked to neuronal differentiation in the olfactory epithelium. Cells 11, 298 (2022).

Article PubMed PubMed Central Google Scholar

Piechota, M. et al. Is senescence-associated -galactosidase a marker of neuronal senescence? Oncotarget 7, 8109981109 (2016).

Article PubMed PubMed Central Google Scholar

Jurk, D. et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 11, 9961004 (2012).

Article CAS PubMed Google Scholar

Moreno-Blas, D. et al. Cortical neurons develop a senescence-like phenotype promoted by dysfunctional autophagy. Aging 11, 61756198 (2019).

Article CAS PubMed PubMed Central Google Scholar

Bigagli, E. et al. Long-term neuroglial cocultures as a brain aging model: hallmarks of senescence, microRNA expression profiles, and comparison with in vivo models. J. Gerontol. A Biol. Sci. Med. Sci. 71, 5060 (2016).

Article CAS PubMed Google Scholar

Kang, H. T., Lee, K. B., Kim, S. Y., Choi, H. R. & Park, S. C. Autophagy impairment induces premature senescence in primary human fibroblasts. PLoS ONE 6, e23367 (2011).

Article ADS CAS PubMed PubMed Central Google Scholar

Wong, A., Kieu, T. & Robbins, P. D. The Ercc1/ mouse model of accelerated senescence and aging for identification and testing of novel senotherapeutic interventions. Aging 12, 2448124483 (2020).

Article CAS PubMed PubMed Central Google Scholar

Yousefzadeh, M. J. et al. Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice. Aging Cell 19, e13094 (2020).

Article CAS PubMed PubMed Central Google Scholar

de Waard, M. C. et al. Age-related motor neuron degeneration in DNA repair-deficient Ercc1 mice. Acta Neuropathol. 120, 461475 (2010).

Article CAS PubMed PubMed Central Google Scholar

Doll, M. E. et al. Broad segmental progeroid changes in short-lived Ercc1/7 mice. Pathobiol. Aging Age Relat. Dis. 1, 10.3402/pba.v1i0.7219(2011).

Sepe, S. et al. Inefficient DNA repair Is an aging-related modifier of Parkinsons disease. Cell Rep. 15, 18661875 (2016).

Article CAS PubMed PubMed Central Google Scholar

Pachajoa, H. et al. HutchinsonGilford progeria syndrome: clinical and molecular characterization. Appl. Clin. Genet. 13, 159164 (2020).

Article CAS PubMed PubMed Central Google Scholar

Machiela, E. et al. The interaction of aging and cellular stress contributes to pathogenesis in mouse and human huntington disease neurons. Front Aging Neurosci. 12, 524369 (2020).

Article CAS PubMed PubMed Central Google Scholar

Baek, J. H. et al. Expression of progerin in aging mouse brains reveals structural nuclear abnormalities without detectible significant alterations in gene expression, hippocampal stem cells or behavior. Hum. Mol. Genet. 24, 13051321 (2015).

Article CAS PubMed Google Scholar

Arendt, T., Rdel, L., Grtner, U. & Holzer, M. Expression of the cyclin-dependent kinase inhibitor p16 in Alzheimers disease. Neuroreport 7, 30473049 (1996).

Article CAS PubMed Google Scholar

McShea, A., Harris, P. L., Webster, K. R., Wahl, A. F. & Smith, M. A. Abnormal expression of the cell cycle regulators P16 and CDK4 in Alzheimers disease. Am. J. Pathol. 150, 19331939 (1997).

CAS PubMed PubMed Central Google Scholar

Wang, B. et al. An inducible p21-Cre mouse model to monitor and manipulate p21-highly-expressing senescent cells in vivo. Nat. Aging 1, 962973 (2021).

Article PubMed PubMed Central Google Scholar

Hu, W. et al. Direct conversion of normal and Alzheimers disease human fibroblasts into neuronal cells by small molecules. Cell Stem Cell 17, 204212 (2015).

Article CAS PubMed Google Scholar

Mertens, J. et al. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell 17, 705718 (2015).

Article CAS PubMed PubMed Central Google Scholar

Yoo, A. S. et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476, 228231 (2011).

Article ADS CAS PubMed PubMed Central Google Scholar

Capano, L. S. et al. Recapitulation of endogenous 4R tau expression and formation of insoluble tau in directly reprogrammed human neurons. Cell Stem Cell 29, 918932 (2022).

Article CAS PubMed PubMed Central Google Scholar

Zhang, C. et al. Establishing RNAge to score cellular aging and rejuvenation paradigms and identify novel age-modulating compounds. Preprint at bioRxiv https://doi.org/10.1101/2023.07.03.547539 (2023).

Sun, Z. et al. Endogenous recapitulation of Alzheimers disease neuropathology through human 3D direct neuronal reprogramming. Preprint at bioRxiv https://doi.org/10.1101/2023.05.24.542155 (2023).

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

See original here:
Lessons from inducible pluripotent stem cell models on neuronal senescence in aging and neurodegeneration - Nature.com

Company Trying to Resurrect a Mammoth Makes a Stem Cell Breakthrough – Gizmodo

Colossal Biosciences, which calls itself the worlds first de-extinction company, has created stem cells it thinks will hasten the companys marquee goal of resurrecting the woolly mammoth. The teams research describing the accomplishment will be hosted on the preprint server bioRxiv.

What Drew John Boyega Back Into Sci-Fi? | io9 Interview

The cells are induced pluripotent stem cells (iPSC), a type of cell that can be reprogrammed to develop into any other type of cell. The cells are especially useful in bioengineering, for their applications in cell development, therapy, and transferring genetic information across species. Colossals new iPSCs are the first engineered elephant cells converted into an embryonic state, a useful development if youre in pursuit of a woolly mammoth. Or rather, an animal that looks like a woolly mammoth.

In the past, a multitude of attempts to generate elephant iPSCs have not been fruitful. Elephants are a very special species and we have only just begun to scratch the surface of their fundamental biology, said Eriona Hysolli, who heads up Colossals biological sciences team, in a statement. The Colossal mammoth team persisted quite successfully as this progress is invaluable for the future of elephant assisted reproductive technologies as well as advanced cellular modeling of mammoth phenotypes.

According to the Colossal release, the new stem cells were able to differentiate into the three germ layers that result in every cell type. It opens the door to establishing connections between genes and traits for both modern and extinct relativesincluding resistance to environmental extremes and pathogens, said George Church, a geneticist and co-founder of Colossal, in a press release.

The animals Colossal hopes to produce will be Asian elephants (E. maximus), genetically engineered to be resistant to the cold and, most notably, covered in shaggy hair la woolly mammoth, their extinct cousin. Colossal also has plans to produce approximate (or proxy) species of the Tasmanian tiger or thylacine, which went extinct around 1936, and the dodo, a flightless bird native to Mauritius, which was gone by 1681. Other companiesnamely Revive & Restorehave similar aims with other species, including the heath hen and passenger pigeon.

A proxy species isnt truly the old creature brought back to life. As described in a 2016 report by the International Union for Conservation of Natures Species Survival Commission, Proxy is used here to mean a substitute that would represent in some sense (e.g. phenotypically, behaviourally, ecologically) another entity the extinct form. The group added that Proxy is preferred to facsimile, which implies creation of an exact copy.

One expert who spoke to Gizmodo previously referred to the end-goals of these companies as something out of Lovecraft and the elephantine effort as a simulacrum that has no phylogenetic relationship with actual mammoths.

Its not just a question of having biological material from an extinct animal. Researchers exploring the possibility of resurrecting the Christmas Island rat found that some genetics were simply lost to time, in spite of the amount that could be gleaned from historic tissues and its nearest extant relatives. One member of the team told Gizmodo that We arent actually planning to do it, as probably the world doesnt need any more rats, and probably the money it would take to do the best job possible could be spent on better things, e.g., conserving living things. (That researcher is now a member of Colossals advisory board.) Nevertheless, the production of elephant iPSCs is a step toward producing these proxy animals, an aim that many scientists see as likely but fewer see as useful.

Once Colossal produces a herd of proxy mammoths, its intention is to decelerate the melting of the permafrost by loosing the animals on a swath of Siberia. Ultimately, Colossal says, the mammoth steppethe ancient ecosystem in which the giant proboscideans roamedcould be restored, helping fight climate change and pushing new technologies in gene editing in the process, helping extant elephants, which face their own survival threats.

But other technological breakthroughs will be necessary to make any of that possible. As noted by Nature, Church intends to use artificial elephant wombs to produce the proxy mammoths, so as to not require Asian elephant surrogates. Asian elephants are an endangered species; to use them as surrogates for proxy mammoths would be the cherry-on-top of an ethical dilemma sundae.

Were still a long way off from Colossals ultimate goals, but this recent achievement is a significant one, and a reminder that these de-extinction efforts involve serious science.

See the original post here:
Company Trying to Resurrect a Mammoth Makes a Stem Cell Breakthrough - Gizmodo

A human embryo model mimics early development and blood-cell formation – Nature.com

Human embryos are extremely difficult to study. This lack of samples limits our understanding of crucial developmental stages, such as the early formation of blood cells. A stem-cell-based model closely captures the development of human embryonic and key extra-embryonic tissues after implantation, as well as the formation of early blood cells.

See the original post here:
A human embryo model mimics early development and blood-cell formation - Nature.com

Breaking through new research barriers with iPSC technology – News-Medical.Net

In this interview, we speak to Keith Olson and Coby Carlson from FUJIFILMCellular Dynamics about their iPSC technology and how it is making breakthroughs in blood-brain barrier research.

Coby Carlson: My name is Coby Carlson, and I lead the Applications Team at FUJIFILMCellular Dynamics. Our team is separate from the R&D scientists, staff, and experts focusing on stem cell culture and differentiations to create unique, specialized cell types. Our role is to test the functionality of these cells, demonstrate their performance on various platform technologies, and develop applications.

Keith Olson: My name is Keith Olson, and I am the Executive Vice President for Commercial Operations at FUJIFILM Cellular Dynamics.

Coby Carlson: Fujifilms core technologies include reprogramming, engineering, and iPSC culture and differentiation to create unique cell types. Our mission is to bring these technologies to an industrial scale. The technology was first invented over 15 years ago, and since then, we have continuously improved and optimized it to launch it on a large scale. Our goal is to make different cell types accessible all over the world.

Keith Olson: I believe that we were pioneers of iPSC technology in the United States. We successfully brought it to market, commercialized it, and built a business around it. Our reputation is now based on our core expertise, quality, and ability to manufacture at scale for our clients in the pharmaceutical, biotech, and academic sectors.

Image Credit:metamorworks/Shutterstock.com

Coby Carlson: The primary focus of people working with iPSC technology is toxicology/safety pharm, disease modeling, and cell therapy. Safety is crucial because we can generate cardiomyocytes from iPS cells, and these cells spontaneously beat in a dish while responding to known cardiotoxic molecules.

To measure the safety and effectiveness of drugs in a dish, we use iPSC-derived cardiomyocytes in safety toxicology. We have also participated in the CiPA(Comprehensive in Vitro Proarrhythmia Aassay) initiative to standardize this process across various labs globally. The recent FDA Modernization Act has suggested using alternatives to animal testing, making iPSC-derived cardiomyocytes a crucial component of measuring the safety and effectiveness of drugs going forward.

Keith Olson: iPSC delivers a specific gap in the market by providing customers with high-quality human cells. Instead of researching animal models or transformed cell lines, researchers can now use real human cardiomyocytes or neurons for more biologically relevant results.

Coby Carlson: The uniqueness of iPSC technology lies in the fact that there are not many human donors who can offer their brain cells. This technology provides access to different cell types, and we can differentiate highly specialized cells to obtain unique features such as excitatory neurons and microglia. These unique cells are essential for in vitro assays, where we can test them differently than we would in an animal model. For instance, mouse models are different from human models.

Keith Olson: Everything starts from a core iPS cell bank, and we have developed specific differentiation protocols for each terminal phenotype of the cell. Whether it is a cardiomyocyte, neuron, or hepatocyte, we have a differentiation protocol that takes us from the core stem cell bank to a finished product that customers can purchase and use as an assay-ready sample.

Image Credit:Magic mine/Shutterstock.com

Coby Carlson: Researchers and drug developers have long struggled with the blood-brain barrier, a critical cell structure separating the blood from the brain. The barrier keeps out harmful pathogens and toxins while allowing vital nutrients to enter the brain. However, it also prevents drugs from crossing over and treating neurological diseases such as neurodegenerative disorders.

The main challenge has been replicating this barrier in vitro. One of the reasons we pursued iPSC cell technology is that we developed ways to distinguish specialized cell types from the same donor to create the blood-brain barrier. Additionally, we can cryopreserve these cells and generate media that allows them to be functional and reanimated from the thaw, enabling us to test them in an in vitro assay.

Keith Olson: We chose to develop a highly relevant human model for studying the blood-brain barrier for several reasons. The industry bottleneck and challenge in understanding how some large molecules cannot pass through the barrier made it necessary. We needed a system that truly replicated what happens in the human brain. We helped revolutionize the market by making the cell type, which was particularly difficult to cryopreserve and develop as an assay-ready product. We were able to deliver a fully isogenic solution with all cell types, and now customers can build their own real human BBB in an assay plate.

Coby Carlson: Generating the BBB is a significant challenge that requires assembling three different cell types. We validated our model using a Transwell assay, which measures the function of the cells on both the top and bottom of the barrier. However, we recognize that the BBB is a complex system, and microfluidic and organ-on-a-chip technologies offer a unique environment for the cells to function. Combining iPSC technology and OoC technologies will enhance cell function in these systems by providing unique environments with microfluidics and dynamic flow similar to the bloodstream. This will allow us to demonstrate the advantages of these models in a more comprehensive manner.

Keith Olson: The system is perfect for 3D modeling because it is a 3D system. It is a multi-layer, multi-cell-type organ or part of an organ, and using it as an organoid or spheroid in a complex model brings you closer to that model system. Building the barrier is crucial, and 3D modeling is the best way to achieve this. Our released product is flexible enough for clients to conduct assays in a Transwell plate or as a spheroid. They can even use more sophisticated organ-on-a-chip systems to create a comprehensive model for this biology.

Coby Carlson: When we started the company, we differentiated cells from iPS lines to create highly purified neurons. While they were unique and visually pleasing, we discovered that some of their functionality was lacking. Ten years later, we have developed various products, intending to combine them to create multicellular cultures, not just in 2D systems but also in 3D systems.

Our approach involves mixing different cell types to increase complexity and biological relevance, which we believe will help improve testing and facilitate the development of new therapies and the identification of new compounds.

Keith Olson: In neurobiology, it is well-established that a human system is essential. Animal model systems simply do not replicate human biology, and studying true human biology requires a 3D system. We are not flat figures but 3D beings, and building a real neural system allows us to observe neural connections, communication, and the impact of glia on neurons, studying how cells interact as they would in the body. This is where the true value lies in what we are delivering.

Image Credit:Naumova Marina/Shutterstock.com

Coby Carlson: As a technology, the use of human iPS drive cell types is still in its infancy, with significant advances still being made. We believe that some major discoveries will be made by using this technology, whether that be by us or others. This technology has the potential to facilitate the discovery of new treatments, and we are excited about its future potential.

Keith Olson: I think anything that any of our competitors, colleagues, friends, and neighbors are doing in this space will help push the field forward. The more vendors and researchers working on creating relevant model systems for humans, the better. With the FDA Modernization Act and the desire for better test systems and models, everyone working together will likely result in better systems and, hopefully, faster, cheaper, and easier drug discovery for companies.

Coby Carlson: The SLAS conference is significant as it brings together various customers, collaborators, friends, and former colleagues operating in this field. The past years haveseen challenges due to the pandemic and remote work, with limited ability to collaborate effectively. The opportunity to meet face-to-face is essential when pushing these models forward and persuading companies and groups to invest significant time and resources. Having these conversations in person allows for quick decision-making and improves the efficiency of the process.

Keith Olson: The SLAS conference seems to have finally returned to normality after the pandemic. The number of attendees and vendors is impressive, and the human-to-human interaction is just unbeatable, especially when it comes to answering queries and gaining an understanding of something first-hand. Our focus remains on highlighting our entire portfolio, but we also showcased some new systems, such as the iCell Macrophage, which is the first of its kind.

Additionally, we continued to promote our BBB system and highlighted other 3D model systems we have developed, including in the neuro and cardio spheres. Overall, it was an opportunity to build awareness for our company and its offerings.

Industrial Scale Cell Engineering - FUJIFILM Cellular Dynamics at SLAS 2023Play

Coby Carlson: Reviews and testimonials are popular among our customers, who are primarily scientists requiring supporting evidence. We understand that content is essential, so we offer it in various formats, including small bites, digital graphical abstracts, and detailed protocols. We aim to cater to the diverse needs of our customers, providing them with the relevant information to make informed decisions about our products.

Keith Olson: We all learned a valuable lesson during the pandemic that content is king. As a result, we had to adapt and utilize various means such as websites, LinkedIn, and other portals to disseminate information to our customers and clients. Due to the absence of trade shows, these platforms were our only options. However, now that trade shows are seemingly back in full swing and everyone is attending, it presents a more effective way to communicate to a large audience at once, which is always more enjoyable.

Coby Carlson: While we did not discuss it much, our cell therapy is where the truly exciting developments will take place and capture the general publics attention. For instance, curing blindness by using retinal cells is something that could happen in our lifetime. Although I cannot speculate on the exact timeline, I believe it is inevitable. Our in vitro work behind the scenes in drug discovery and other aspects aligns well with this goal, as the same cells used in our research could be used as drugs in the future.

Keith Olson: An exciting prospect on the horizon is the potential replacement of animal model systems with validated human models within the next year or five years. If this were to happen, and the FDA, pharma companies, and biotechs were on board, it would be a major advancement for everyone. This could result in faster, cheaper, and more efficient processes. One of the main challenges in this field is the high rate of failure, and any improvements made in this area could potentially save billions of dollars for the industry.

Coby Carlson: We always strive to advance our field, evaluating new technologies and creating new products. We want to expand our collaborations and work with others to achieve our scientific goals. While we can conduct R&D, we recognize that partnering with other experts, sharing information, and collaborating can lead to even greater success.

Keith Olson: Our company is constantly exploring new cell backgrounds, and we are committed to launching two to three new cell types each year. Currently, we are focused on introducing three new cell types, all of which will be launched by June or July of this year. Simultaneously, we have begun developing the next three cell types for 2024. Our goal is to continue expanding our offerings, and we hope to return next year with news of three entirely new cell lines.

Keith Olson is the senior commercial executive for FUJIFILM Cellular Dynamics, where we have developed the market-leading portfolio of differentiated human cells derived from iPSC.

Keith has previously served in leadership roles for Life Science products at Corning Inc, Life Technologies, DiscoveRx and Cellomics, and has launched over 700 Life Science products in his career. Keith received his bachelors degree in Molecular Cell Biology from Carnegie-Mellon University and his Ph.D. in the same from the University of Rochester.

Coby Carlson is the Director of Applications Development at FUJIFILM Cellular Dynamics where his team focuses on advancing the use of human iPSC-derived cell types for drug screening, toxicity testing, and disease modeling.

He joined the company in 2012 and has extensive experience developing applications with iCell products, characterizing their functional performance on various technology platforms, and building co-culture systems in both 2D and 3D format. Coby received his PhD from the Univ. of Utah followed by a postdoctoral fellowship at UW-Madison.

Read the rest here:
Breaking through new research barriers with iPSC technology - News-Medical.Net

A low-cost device to make cell therapy safer – Tech Explorist

In cell therapy, clinicians reprogram some skin or blood cells from patients to create induced pluripotent stem cells. They coax these stem cells to transform into progenitor cells for treating spinal cord injury. These progenitors are then transplanted back into the patient to regenerate part of the injured spinal cord. However, pluripotent stem cells that dont entirely change into progenitors can form tumors.

Scientists at MIT and the Singapore-MIT Alliance for Research and Technology have developed a tiny device to improve cell therapy treatments with more excellent safety and effectiveness. They developed a microfluidic cell sorter to remove undifferentiated cells without damaging fully-formed progenitor cells.

This newly developed device can sort more than 3 million cells per minute without special chemicals. In the study, scientists found that combining many devices can sort more than 500 million cells per minute.

Pluripotent stem cells were generally larger than the progenitor cells derived from them. It happens because pluripotent stem cells have many genes that havent been switched off in their nucleus. As these cells specialize in specific functions, they suppress many genes that are no longer required, hence shrinking the nucleus. The microfluidic device leverages this size difference to sort the cells.

The plastic chip contains tiny channels that create an inlet for cells to enter, a spiral pathway, and four outlets where cells of different sizes are collected. When cells pass through the spiral at high speeds, various forces, including centrifugal forces, push them around. These forces help gather the cells at a specific point in the fluid stream based on their size, effectively separating them into different outlets.

The researchers discovered they could enhance the sorters performance by running it twice. First, they operate it at a lower speed, causing more giant cells to stick to the walls while smaller cells are sorted out. Then, they run it faster to separate the larger cells.

The device works similarly to a centrifuge, but it doesnt need human intervention to collect the sorted cells.

The device could remove almost 50% of larger cells in one pass. Whats more, the device doesnt use any filtration. The limitations with filters are that they become clogged or break down over time so that a filter-free device can be used for much longer.

Having demonstrated success on a small scale, the researchers are now moving on to larger studies and animal models to determine if the purified cells work better when introduced into living organisms.

Journal Reference:

Read this article:
A low-cost device to make cell therapy safer - Tech Explorist