Fact-checked by Grok 2 weeks ago

Reprogramming

Cellular reprogramming refers to the process of converting differentiated somatic cells into induced pluripotent stem cells (iPSCs) by erasing epigenetic marks and activating pluripotency genes, thereby reversing cellular differentiation to a stem cell-like state. This technique was pioneered in 2006 by Shinya Yamanaka and colleagues, who identified four transcription factors—Oct4, Sox2, Klf4, and c-Myc (collectively known as OSKM)—sufficient to reprogram mouse embryonic fibroblasts into iPSCs capable of contributing to all cell lineages, including germline transmission. Extended to human cells in 2007 using similar factors on dermal fibroblasts, the method enabled generation of patient-specific iPSCs for disease modeling and potential autologous therapies, circumventing ethical issues of embryonic stem cell derivation. Key mechanisms involve dynamic epigenetic remodeling, including DNA demethylation at pluripotency loci and histone modifications that facilitate a metastable intermediate state before stable pluripotency acquisition. While transformative for regenerative medicine, with Yamanaka receiving the 2012 Nobel Prize in Physiology or Medicine, reprogramming faces challenges like low efficiency (typically 0.01-0.1%), incomplete epigenetic erasure leading to memory retention, and oncogenic risks from factors like c-Myc, prompting ongoing research into non-integrating delivery methods and partial reprogramming strategies to mitigate tumorigenesis while achieving rejuvenation.

Biological Contexts

Embryonic Development

In mammalian embryonic development, epigenetic reprogramming initiates immediately upon fertilization, erasing most marks from the parental s to establish totipotency in the . This process involves a genome-wide reduction in (5mC) levels, reaching a minimum at the stage before during implantation. The reprogramming is essential for zygotic (ZGA), which occurs around the 2- to 8-cell stage in mice and later in humans, enabling embryonic independent of maternal transcripts. The demethylation exhibits asymmetry between parental genomes. In the paternal , active demethylation predominates, driven by the TET3 enzyme that oxidizes 5mC to (5hmC), followed by dilution or further processing via . Conversely, the maternal primarily undergoes passive demethylation through exclusion of maintenance methyltransferase during replication, leading to progressive loss over cell divisions. Studies in mice demonstrate that paternal 5mC levels drop by approximately 80-90% within hours post-fertilization, while maternal levels decline more gradually. Exceptions to global demethylation include differentially methylated regions (DMRs) at imprinted loci, which resist erasure to preserve parent-of-origin-specific expression patterns critical for . Disruption of this reprogramming, such as in mice, impairs ZGA and embryonic viability, underscoring its causal role in early . Accompanying epigenetic changes involve variant exchanges, such as H3.3 incorporation, and opening to facilitate transcriptional priming. In humans, single-cell epigenomic reveals similar , with implications for assisted reproductive technologies where perturbations can lead to developmental abnormalities.

Learning and Memory

Learning and memory processes in the involve epigenetic modifications that dynamically alter patterns in neurons, effectively reprogramming the landscape to encode and stabilize experiences. , a key epigenetic mark, serves as a mechanism for storage by influencing genes in regions such as the and . These changes occur rapidly following learning tasks, with demethylation activating genes required for and hypermethylation repressing others to prevent interference. In contextual fear conditioning experiments with rodents, training induces specific DNA methylation alterations in plasticity-related genes like BDNF and Egr1, which correlate with memory acquisition and persistence up to 24 hours post-training. Enzymes such as DNA methyltransferases (DNMTs), including DNMT3a, catalyze these methylation events in the hippocampus, where their inhibition impairs long-term memory formation without affecting short-term recall. Conversely, ten-eleven translocation (TET) proteins facilitate active demethylation, enabling the expression of genes critical for engram stabilization in neuronal ensembles. Histone modifications complement DNA methylation in this reprogramming, with acetylation promoting open chromatin for transcription during memory encoding, while deacetylation contributes to the maintenance phase. Studies show that inhibiting histone deacetylases (HDACs) enhances memory retention, underscoring the reversible nature of these epigenetic programs that balance plasticity and stability.00433-8) In Drosophila models, the histone methyltransferase EHMT regulates an epigenetic program involving classic learning genes, linking chromatin dynamics to cognitive outcomes across species. Recent advances indicate that partial cellular reprogramming of neurons can rejuvenate age-related declines in , restoring youthful epigenetic states and improving performance in aged mice through controlled expression of factors like OSK (Oct4, , ). This approach highlights reprogramming's potential to modulate learning circuits, though it primarily targets rejuvenation rather than basal formation. Overall, these mechanisms ensure that epigenetic reprogramming provides a molecular basis for enduring behavioral adaptations, with disruptions linked to disorders like where aberrant patterns impair .

Aging and Rejuvenation

Cellular reprogramming targets aging by reversing epigenetic alterations that accumulate over time, such as changes tracked by epigenetic clocks like the Horvath clock. Partial reprogramming, involving transient expression of Yamanaka factors (Oct4, , , and optionally c-Myc, or OSKM), resets these marks to a youthful state while preserving cellular identity, unlike full reprogramming to induced pluripotent stem cells (iPSCs) which erases . This approach addresses causal epigenetic noise and loss of information proposed as drivers of aging phenotypes.01570-7) In models, partial reprogramming has demonstrated functional rejuvenation. A 2020 study expressed OSKM in retinal ganglion cells of aged and glaucomatous mice, restoring youthful transcriptomic profiles, regeneration, and , with treated mice regaining near-normal within weeks. Similarly, cyclic OSK expression (excluding c-Myc to reduce tumorigenicity) in progeroid Zmpste24-/- mice extended median lifespan by approximately 30%, improved body weight, fur condition, and reduced , while lowering epigenetic age by over 50% in multiple tissues. These effects correlated with decreased markers and enhanced tissue repair, though full lifespan extension in wild-type mice remains under investigation. Chemical alternatives to genetic Yamanaka factors have accelerated protocols. In 2023, six chemical cocktails were identified that, applied for less than seven days to human fibroblasts, reversed transcriptomic age by 2-3 years per Horvath clock estimates, restoring youthful without compromising cell identity or inducing pluripotency. In vivo, such partial reprogramming ameliorated age-related molecular changes in physiologically aging mice, including reduced and improved metabolic profiles. A 2025 preprint reported a single-gene , SB000, rivaling OSKM efficacy across germ layers in cellular assays, suggesting streamlined future therapies. Challenges include risks of tumorigenesis from prolonged factor expression and incomplete reversal of all aging hallmarks, such as proteostasis decline. While mouse data indicate causality between epigenetic reprogramming and phenotypic rejuvenation, human applications are preclinical, with epigenetic clock reversals observed in cellular models but not yet in vivo trials. Ongoing research prioritizes safer, non-integrative delivery like gene therapy or small molecules to translate these findings.

Molecular Mechanisms

Epigenetic Modifications

Epigenetic modifications encompass heritable changes in without altering the underlying DNA sequence, primarily through , histone post-translational modifications, and , which collectively govern cellular reprogramming by modulating accessibility and transcriptional programs. In induced pluripotency, these modifications enable the transition from to pluripotent states by erasing lineage-specific marks and reinstating embryonic-like epigenetic landscapes. DNA methylation, characterized by the addition of methyl groups to residues predominantly at CpG dinucleotides, serves as a key repressive mechanism in differentiated cells, silencing pluripotency genes such as Nanog and Oct4 through promoter hypermethylation. During reprogramming with Yamanaka factors (Oct4, , , c-Myc), demethylation occurs primarily in late stages, aligning with the acquisition of embryonic stem cell-like identity, where family enzymes oxidize (5mC) to (5hmC), facilitating passive loss via replication-dependent dilution or active . This process is rate-limiting, with incomplete demethylation at pluripotency loci acting as a barrier; supplementation with enhances TET activity and reprogramming efficiency by up to tenfold in some protocols. Hypermethylation gains, conversely, accumulate gradually at non-pluripotent sites, stabilizing the new identity. Histone modifications provide dynamic regulation, with activating marks like H3K4 deposited early by reprogramming factors to prime enhancers and promoters for transcription, often preceding . Repressive marks, including in and mediated by Polycomb repressive complex 2 (PRC2), pose significant barriers; their removal via demethylases such as UTX (for ) or inhibition of methyltransferases like SUV39H1 (for ) accelerates reprogramming kinetics and boosts colony formation rates. Bivalent domains, bearing both and , resolve during reprogramming to activate developmental regulators while repressing somatic s. inhibitors, like valproic acid, further enhance by promoting , increasing iPSC generation efficiency. In developmental contexts, such as zygotic genome activation, global epigenetic erasure involves rapid TET3-dependent demethylation post-fertilization, mirroring mechanisms in where Tet3 deficiency impairs reprogramming. Additional layers, including non-coding RNAs (e.g., miR-302 cluster targeting repressors) and ATP-dependent remodelers, cooperate to dismantle somatic . Persistent epigenetic memory from donors, such as incomplete dissolution, underlies inefficiencies in and iPSC , underscoring the need for targeted .

Key Enzymes and Factors

The core transcription factors driving (iPSC) reprogramming, known as the Yamanaka factors, consist of Oct4 (Pou5f1), , , and c-Myc (OSKM). These factors, when ectopically expressed in cells, initiate a cascade that overrides lineage-specific and reactivates pluripotency networks. Oct4 and form a heterodimer that binds enhancer regions of pluripotency genes, while and c-Myc facilitate opening and proliferation, respectively. Epigenetic enzymes play crucial roles in erasing memory during reprogramming. Ten-eleven translocation () enzymes, particularly TET1 and TET2, catalyze the oxidation of (5mC) to (5hmC), promoting active at pluripotency loci. This process cooperates with OSKM factors to enable gene reactivation, as TETs are recruited to target sites by pluripotency transcription factors like Nanog. DNA methyltransferases (DNMTs), including for maintenance methylation and DNMT3A/B for methylation, oppose reprogramming by preserving methylation patterns. Successful reprogramming requires downregulation or inhibition of DNMT activity, often achieved through TET-mediated demethylation or replication-dependent dilution. Inhibition of DNMTs enhances reprogramming efficiency, underscoring their barrier role. Other factors, such as histone deacetylases (HDACs), contribute by modulating chromatin accessibility, with HDAC inhibitors boosting OSKM-induced reprogramming. However, the primary drivers remain the OSKM transcription factors and TET-DNMT axis for epigenetic reconfiguration.

Transcriptional Regulation

Transcriptional regulation in cellular reprogramming centers on the ectopic expression of core transcription factors, notably the Yamanaka factors—Oct4, Sox2, Klf4, and c-Myc (OSKM)—discovered by Shinya Yamanaka's team in 2006, which reprogram somatic cells to induced pluripotency by overriding somatic gene networks and establishing a pluripotent transcriptional program. These factors collaborate to repress lineage-specific enhancers while activating pluripotency-associated enhancers and promoters, initiating a self-reinforcing endogenous regulatory circuit involving genes like Nanog and Sall4. OSK (Oct4, Sox2, Klf4) predominantly target enhancers, recruiting and redirecting somatic transcription factors such as AP-1 and C/EBP away from their native sites, whereas c-Myc exerts broader effects at promoters to facilitate proliferation and global transcriptional amplification. The process unfolds in temporally distinct phases marked by dynamic shifts. Early reprogramming features stochastic binding of OSK to somatic and transient enhancers, driving mesenchymal-to-epithelial (MET) via downregulation of mesenchymal genes (e.g., Snai1, Zeb1) and upregulation of epithelial markers (e.g., E-cadherin), which is essential for progression but constitutes a barrier in fibroblasts. Intermediate stages involve partial activation of early pluripotency genes like Utf1 and Esrrb, with opening at select loci; late phases to deterministic activation of the full pluripotency network, including core factors like endogenous Oct4 and Nanog, stabilizing the reprogrammed state. Transcriptional pausing at promoters of pluripotency genes represents a key rate-limiting step, where OSKM recruit P-TEFb to release paused , enabling efficient elongation and gene activation. Pioneer activity of and enables initial access to compacted , often in cooperation with to enhance long-range interactions and topological domain connectivity, thereby facilitating enhancer-promoter looping for target gene activation. Dosage of these factors critically modulates outcomes: suboptimal levels increase heterogeneity and inefficiency, while balanced ratios—such as equimolar OSKM—optimize enhancer occupancy and reduce stochastic barriers, as evidenced by single-cell analyses showing dose-dependent trajectories in reprogramming intermediates. Despite advances, residual somatic transcriptional memory persists in iPSCs, influencing bias and underscoring incomplete erasure of original identities. Overall, transcriptional reprogramming integrates , rewiring, and pause release to achieve cell fate conversion, with efficiencies typically below 1% in standard protocols due to these regulatory hurdles.

Historical Development

Early Experiments in Nuclear Transfer and Fusion


The earliest experiments in nuclear transfer, precursors to modern somatic cell nuclear transfer (SCNT), were conducted by Robert Briggs and Thomas J. King in 1952 using embryos of the frog Rana pipiens. They transplanted nuclei from blastula-stage donor cells into enucleated eggs, achieving development to tadpole stages, though success rates were low and further progression to fertile adults was not observed. These experiments demonstrated that early embryonic nuclei could direct development but highlighted challenges with differentiated nuclei, including incomplete reprogramming due to nuclear-cytoplasmic incompatibilities. John B. Gurdon advanced these techniques in the late 1950s and early 1960s using Xenopus laevis eggs, which allowed ultraviolet irradiation for enucleation without mechanical damage. In 1962, Gurdon successfully cloned tadpoles and, through serial nuclear transfers, produced fertile adult frogs from nuclei of differentiated intestinal epithelial cells of feeding tadpoles. Out of approximately 270 reconstructions with differentiated nuclei, only about 1-2% developed into normal frogs, underscoring the inefficiency but confirming that somatic nuclei retain totipotency when exposed to egg cytoplasm, which actively reprograms structure and . This work established the principle of nuclear reprogramming, showing that does not involve irreversible genetic loss but reversible epigenetic changes. Parallel early experiments in , pioneered by Harris and colleagues in the mid-1960s, provided complementary evidence for reprogramming. In 1965, Harris and J. F. Watkins used inactivated Sendai virus to fuse human and mouse , creating stable lines that exhibited selective and , such as the reactivation of inactive X chromosomes. These heterokaryons revealed cytoplasmic factors capable of influencing nuclear across species barriers, with embryonic or pluripotent cytoplasms dominating to suppress differentiated traits. By the early , fusions between and embryonal demonstrated rapid reprogramming, including pluripotency marker expression and tumorigenic potential in , further illustrating the plasticity of genomes. Such findings supported the existence of reprogramming factors, paving the way for understanding epigenetic dominance in .

Yamanaka's Induced Pluripotency Breakthrough

In August 2006, and colleagues at reported the generation of induced pluripotent stem cells (iPSCs) from mouse embryonic and adult fibroblasts through the introduction of four specific transcription factors: Oct3/4, , , and c-Myc.00976-7) These factors were identified by screening 24 candidate genes highly expressed in embryonic stem cells (ESCs), with combinatorial retroviral transduction enabling reprogramming without or techniques.00976-7) The resulting iPSCs displayed ESC-like morphology, rates, and profiles, including of endogenous pluripotency genes and of somatic markers.00976-7) Pluripotency was rigorously validated through differentiation into derivatives of all three germ layers, teratoma formation in immunocompromised mice, and contribution to viable chimeric mice with germline transmission, confirming functional equivalence to ESCs.00976-7) Reprogramming efficiency was low, approximately 0.01-0.1% of transduced cells, and required the oncogene c-Myc, though later variants omitted it at the cost of reduced efficiency.00976-7) This method bypassed the ethical concerns associated with deriving ESCs from embryos, offering a renewable source of patient-specific pluripotent cells for and potential . In November 2007, Yamanaka's team extended the protocol to human adult dermal fibroblasts, using the same four factors delivered via retroviruses, yielding human iPSCs indistinguishable from human ESCs in pluripotency markers and potential.01471-7) Human iPSCs formed teratomas containing tissues from , , and , though chimeras were not feasible due to ethical constraints.01471-7) The discovery earned Yamanaka the 2012 in Physiology or Medicine, shared with , for demonstrating that mature somatic cells could be reprogrammed to a pluripotent by defined factors, fundamentally altering understandings of cellular and opening avenues for . Despite initial concerns over retroviral integration risks, such as mutagenesis from c-Myc, the approach spurred rapid advancements in non-integrative reprogramming methods.

Post-2006 Advances

In November 2007, Shinya Yamanaka's team reported the generation of human induced pluripotent stem (iPS) cells from dermal fibroblasts using the same four transcription factors—Oct4, , Klf4, and c-Myc—as in the mouse model, achieving pluripotency verified by formation and contribution to chimeric mice.01471-7) Independently, James Thomson's group derived human iPS cells using Oct4, , Nanog, and Lin28, avoiding oncogenic c-Myc to reduce tumorigenicity risks. These breakthroughs extended reprogramming to human cells, enabling patient-specific models while highlighting challenges like low efficiency (around 0.01-0.1%) and viral integration-induced mutations. Subsequent refinements addressed safety and efficiency. In 2008, Rudolf Jaenisch's laboratory produced mouse cells without c-Myc, demonstrating comparable pluripotency and reduced tumor incidence upon transplantation. By 2009, further optimizations yielded integration-free methods, including protein of reprogramming factors, as shown by Qiang Zhou's group, which reprogrammed mouse fibroblasts using cell-permeable Oct4, , Klf4, and c-Myc proteins linked to poly-arginine, bypassing genetic material altogether.00183-0) Non-integrating viral approaches, such as Sendai virus vectors reported in 2011, improved human generation by enabling transient factor expression without genomic insertion. These advances increased clinical viability, with efficiencies rising to 0.1-1% in optimized protocols. Beyond full pluripotency, direct lineage emerged as a . In 2010, Marius Wernbürger and colleagues converted mouse fibroblasts into functional neurons using three neural-lineage-specific factors—Ascl1, Brn2, and Myt1l—without passing through an pluripotent intermediate, achieving neuronal and electrophysiologic maturity within 2-3 weeks. This approach extended to other lineages, such as fibroblasts to cardiomyocytes (2010, Ieda et al.) and hepatocytes, offering targeted therapies for repair. Partial reprogramming, avoiding full , gained traction for ; in 2016, Manuel Serrano's team used doxycycline-inducible OSK factors (Oct4, , ) in progeric mice, restoring epigenetic youth markers, enhancing repair, and extending median lifespan by 30% without tumorigenesis. Chemical reprogramming marked another milestone, leveraging small molecules to mimic factor effects. In 2013, early cocktails partially reprogrammed cells, but full chemical induction of iPS cells was achieved in 2015 by Jing Qu's group using a seven-compound mix (including VC6TF, CHIR99021, and RepSox) over 50 days, activating endogenous pluripotency networks without exogenous genes.00291-9) Recent extensions include chemical reprogramming prototypes by 2022, though efficiencies remain low (0.001-0.01%).00345-3) In vivo applications advanced, with 2016 studies demonstrating direct cardiac reprogramming in hearts post-injury via Gata4, Mef2c, and Tbx5, improving function by 20-30%. By 2023, clinical trials using iPS-derived cells for Parkinson's (e.g., Cell Therapeutics) and underscored translation, with Japan's 2014 retina trial as the first iPS-based therapy. These developments prioritized epigenetic barrier dismantling, with TET enzymes and DNA demethylases identified as key mediators.

Reprogramming Processes

Initiation Phase

The initiation phase of cellular reprogramming, particularly in the generation of induced pluripotent stem cells (iPSCs), encompasses the early stochastic events that begin to dismantle the identity and prime cells for pluripotency. This phase is marked by rapid changes in , including the downregulation of mesenchymal genes and upregulation of epithelial markers, facilitating a mesenchymal-to-epithelial (MET). MET is considered a hallmark early event, driven by transcription factors such as Oct4, , , and c-Myc (OSKM), which initiate binding to target loci despite the persistence of repressive epigenetic marks. During initiation, cells exhibit increased proliferation rates, with upregulation of progression genes occurring as one of the earliest detectable changes, often within the first few days of factor induction. Morphological alterations, such as cell flattening and the formation of small aggregates, become visible around day 2 to 6 in human fibroblasts undergoing doxycycline-inducible OSKM expression. These are highly heterogeneous, with only a subset of cells responding due to activation of early pluripotency genes like Sall4 and Esrrb, while somatic transcriptional programs are partially silenced. Epigenetically, the initiation phase involves preliminary remodeling, including the deposition of active marks like at pluripotency loci and initial mediated by TET enzymes, which oxidize to (5hmC). This demethylation begins at specific CpG sites, facilitating access for reprogramming factors, though global epigenetic barriers such as bivalent domains and high levels remain largely intact, contributing to the inefficiency of this phase. of TET proteins impairs these early changes and blocks progression, underscoring their role in overcoming epigenetic memory. Initiation is distinguished from later phases by its reliance on metabolic shifts, including a temporary glycolytic switch, and the absence of stable pluripotency marker expression like Nanog, which emerges only in subsequent maturation. Studies indicate that while initiation events are relatively permissive, they set the stage for more deterministic progression, with barriers like proliferative arrest overcome through proliferation enhancement. Overall, this phase highlights the probabilistic nature of reprogramming, where early successes in MET and epigenetic priming determine the potential for full pluripotency acquisition.

Maturation Phase

The maturation phase of cellular reprogramming follows the initiation phase and is characterized by the progressive activation of the endogenous pluripotency gene regulatory network, including upregulation of core factors such as Nanog, Sox2, and Sall4, alongside partial erasure of somatic epigenetic marks to approach an embryonic stem cell-like state. During this stage, reprogramming intermediates exhibit increased expression of early pluripotency markers like Utf1 and Dppa2, while beginning to remodel chromatin accessibility and DNA methylation patterns, though full global demethylation remains incomplete until stabilization.30386-2) This phase typically spans days 8–12 in standard Oct4, Sox2, Klf4, and c-Myc (OSKM)-driven fibroblast-to-iPSC protocols in mice, marked by metabolic shifts toward glycolysis and enhanced proliferative capacity. A key barrier in maturation is the inefficient traversal from proliferative, epithelial-like intermediates to cells competent for pluripotency acquisition, with studies identifying this as the primary rate-limiting step rather than , as evidenced by time-lapse imaging showing stalled progression in human fibroblasts despite successful early entry. Epigenetic remodeling during maturation involves active demethylation at pluripotency loci via enzymes and modifications, facilitated by auxiliary factors like Dppa2/4, which promote H2A.X-dependent accessibility changes to suppress programs.30386-2) Sustained exogenous factor expression can hinder late maturation by preventing the upregulation of stabilization-phase markers, underscoring the need for transient activity to enable endogenous network dominance. In human reprogramming, maturation efficiency is further constrained by species-specific epigenetic barriers, such as persistent at certain loci, requiring adjuncts like signaling to drive mesenchymal-to-epithelial transition completion and propel cells forward.00170-0) Variations in maturation dynamics across protocols—e.g., faster progression with chemical cocktails—correlate with accelerated acquisition of ES-like cytoskeletal and transcriptomic features, though incomplete maturation often leads to aberrant propensity in derived iPSCs. Overall, maturation represents a metastable where cells commit irreversibly toward pluripotency, with multi-omic analyses revealing coordinated waves of enhancer activation as a hallmark.

Stabilization Phase

The stabilization phase constitutes the concluding stage of somatic cell reprogramming to induced pluripotency, wherein cells attain a self-sustaining pluripotent state independent of exogenous reprogramming factors. This phase follows the maturation stage, during which initial pluripotency markers are activated but transgene dependency persists, and is characterized by the consolidation of the pluripotency gene regulatory network. Key transcriptional events include the late upregulation (typically after day 9 in mouse fibroblasts) of endogenous pluripotency genes such as , Utf1, Lin28a, Dnmt3l, Dppa2, Dppa3, Dppa4, and , forming a secondary wave driven primarily by endogenous Oct4 and Sox2. Markers of successful stabilization encompass Nanog, Sall4, and endogenous Oct4 expression, signifying the establishment of a transcriptional profile akin to embryonic stem cells. Epigenetically, stabilization involves dynamic remodeling to lock in the pluripotent identity: at promoters of pluripotency loci to facilitate their sustained activation, coupled with methylation at differentiation-associated genes like HoxA10 and Gja8. This process ensures complete repression of the somatic gene program and resistance to differentiation cues. A critical requirement for progression is the silencing or removal of transgenes encoding OSKM (Oct4, , , ) factors, as their continued expression suppresses stabilization markers and impedes transgene independence. Studies demonstrate that premature transgene cessation halts reprogramming, while late withdrawal enables the transition, highlighting distinct regulatory pathways from earlier phases. Failure to achieve stabilization results in partially reprogrammed cells prone to reversion or aberrant , underscoring the phase's role in generating bona fide iPSCs capable of long-term self-renewal and multilineage potential.

Experimental Methods

Somatic cell nuclear transfer (SCNT) reprograms differentiated somatic cells to a totipotent state by inserting the donor nucleus into an enucleated , leveraging the egg's cytoplasmic factors to remodel the somatic epigenome. This technique, first demonstrated in amphibians by in 1962 using frog nuclei transferred to enucleated eggs, achieved mammalian success with the sheep in 1996, where an adult cell nucleus yielded a viable after 277 attempts, highlighting early low efficiency. The SCNT process begins with enucleation of a metaphase II to remove its , followed by injection of a quiescent , often using micromanipulation or electrofusion. Activation via chemical stimuli like ionomycin and induces embryonic development, during which oocyte factors drive rapid epigenetic erasure, including and modifications, to restore totipotency within hours. In reprogramming applications, SCNT-derived blastocysts yield embryonic stem cells (ESCs) with pluripotency verified by formation and contribution, enabling patient-matched cells for therapeutic without viral integration risks inherent in induced pluripotency methods.30300-X) Efficiency remains a primary limitation, with live birth rates typically under 5% in mice and even lower in larger mammals due to incomplete reprogramming, aberrant , and placental defects from persistent somatic epigenetic memory. SCNT-ESCs were first derived in 2013 from fetal somatic cells, with fibroblast success reported shortly after, though yields stayed below 1% without enhancements.30056-X) Recent advances, such as histone demethylase overexpression or HDAC inhibitors, have boosted formation to over 20% in some protocols by facilitating opening and X-chromosome reactivation.30300-X) Compared to (iPSC) generation, SCNT achieves more faithful epigenetic resetting but demands scarce oocytes and raises ethical concerns over destruction.30300-X) Ongoing refinements target pre- and post-implantation barriers to enhance viability for .

Cell Fusion Techniques

techniques reprogram cells by merging them with pluripotent cells, such as embryonic stem cells (ESCs), allowing the nucleus to acquire pluripotency through shared cytoplasmic factors without initial genetic material exchange. This process forms heterokaryons, where multiple nuclei coexist in a common , enabling rapid epigenetic reprogramming of the genome. Pioneered by Helen Blau in the 1980s, early experiments fused human fibroblasts with muscle cells, demonstrating activation of muscle-specific genes in non-muscle nuclei, highlighting cellular plasticity. Common methods include chemical fusion using (PEG), which induces membrane destabilization and merger, and electrofusion, applying electric pulses to align and fuse cells via dielectric breakdown. Viral methods, such as inactivated virus, promote fusion through hemagglutinin-neuraminidase interactions. In reprogramming assays, fusing cells with ESCs activates pluripotency markers like Oct4 within days, with nearly all reprogrammed nuclei undergoing within 24 hours post-fusion. Reprogramming efficiency via exceeds that of some transcription factor-based methods, often achieving pluripotency in over 90% of hybrids under optimized conditions, though resulting cells are typically tetraploid due to nuclear . Mechanisms involve diffusion of reprogramming factors from the pluripotent , triggering demethylation and in the somatic , independent of initially. Blau's 2002 analysis emphasized 's role in revealing early reprogramming events, such as gene activation without . Limitations include hybrid cell instability, potential tumorigenicity from , and challenges in isolating mononucleate reprogrammed cells, restricting therapeutic applications compared to transgene-free alternatives. Despite this, serves as a model for studying reprogramming dynamics, informing factor-based methods by identifying essential cytoplasmic components.

Defined Factor Induction

Defined factor induction reprograms somatic cells to pluripotency through the forced expression of specific transcription factors that activate endogenous pluripotency genes and remodel the epigenetic landscape. This method was pioneered in 2006 when Kazutoshi Takahashi and transduced mouse fibroblasts with retroviral vectors encoding Oct4, , , and c-Myc (OSKM), generating colonies resembling embryonic stem cells capable of contributing to chimeric mice and germline transmission.00976-7) The OSKM factors were selected from 24 candidates known to maintain pluripotency in embryonic stem cells, with combinatorial testing revealing their sufficiency for reprogramming. In the original protocol, reprogramming efficiency was low, with approximately 0.02-0.1% of transduced mouse fibroblasts forming cell colonies after 2-3 weeks, reflecting stochastic reactivation of pluripotency networks amid dominant barriers. Yamanaka's approach extended to fibroblasts in 2007 using the same OSKM factors delivered via retroviruses, yielding cells that expressed pluripotency markers and differentiated into all three layers, though initial efficiencies remained below 0.01%. Retroviral integration posed risks of , prompting subsequent refinements such as excisable vectors and non-integrating alternatives. Delivery methods have evolved to enhance safety and efficiency, including lentiviral vectors for stable expression, Sendai virus for transient non-integrating delivery achieving up to 1% efficiency in optimized human protocols, and mRNA-based transfection avoiding DNA intermediaries altogether. Small molecules targeting epigenetic modifiers, like DNA methyltransferase inhibitors, further boost OSKM-mediated reprogramming by 10-100 fold by alleviating chromatin barriers. Variations omitting c-Myc reduce tumorigenicity but halve efficiency, underscoring the factor's role in proliferation during early phases. These advancements have made defined factor induction the most versatile experimental method for generating patient-specific iPS cells for research and potential therapy.

Applications and Techniques

In Vitro Cell Culture Systems

In vitro cell culture systems facilitate the reprogramming of differentiated cells, such as fibroblasts, into induced pluripotent stem cells (iPSCs) by introducing transcription factors like Oct4, , , and c-Myc via viral vectors or non-integrating methods. These systems maintain cells under controlled conditions that suppress and promote pluripotency acquisition, typically starting with plating on supportive substrates and transitioning to embryonic stem cell-like media. Traditional protocols employ feeder layers of mitotically inactivated mouse embryonic fibroblasts (MEFs) to secrete factors inhibiting and supporting colony expansion, with culture media consisting of DMEM supplemented with 15-20% (FBS), non-essential amino acids, L-glutamine, β-mercaptoethanol, and (LIF) for mouse cells. Human iPSC generation often uses xenogeneic-free alternatives, such as MEF-conditioned media or direct feeder-free setups on Matrigel-coated dishes with mTeSR1 medium containing bFGF. Reprogramming progress is monitored via morphological changes to compact colonies and expression of pluripotency markers like Nanog or Tra-1-60, confirmed by staining or qPCR. Feeder-free and defined culture systems have advanced since the mid-2010s, reducing batch-to-batch variability and enabling scalability; for instance, or Synthemax coatings paired with E8 medium support human iPSC maintenance without animal-derived components. or bioreactor cultures, using aggregates in spinner flasks or microcarriers, allow expansion to billions of cells for therapeutic applications, with yields improved by or small-molecule enhancers like valproic acid. Efficiencies range from 0.001% to 1%, influenced by donor cell age, reprogramming method, and culture optimization. Non-integrative approaches, such as Sendai virus or episomal plasmids, integrated into these systems minimize risks, while chemical cocktails partially replace factors to boost . involves karyotyping, pluripotency assays (e.g., teratoma formation), and epigenetic profiling to ensure stable reprogramming without residual memory.

In Vivo Reprogramming

In vivo reprogramming refers to the direct alteration of cellular identity within a living , typically by introducing exogenous transcription factors or chemical agents to convert differentiated cells into pluripotent states, multipotent progenitors, or alternative lineages such as neurons or cardiomyocytes, without prior extraction and manipulation. This approach aims to harness endogenous cells for tissue repair and regeneration, contrasting with in methods that require and culture. Initial demonstrations of in vivo pluripotency induction occurred in 2013, when transient expression of Yamanaka factors (Oct4, , , c-Myc; OSKM) in mouse cells generated teratomas, confirming totipotency-like potential but highlighting risks of uncontrolled proliferation. Techniques for reprogramming predominantly involve vectors, such as retroviruses or lentiviruses, to deliver lineage-specific cocktails into target tissues. For cardiac repair, intramyocardial injection of Gata4, Mef2c, and Tbx5 (GMT factors) in infarcted mouse hearts post-myocardial infarction converted fibroblasts to cardiomyocyte-like cells, reducing scar size by approximately 50% and improving by 20-30% within weeks, though conversion efficiency remained below 10%. In neural contexts, of NeuroD1 or Ascl1 into reprogrammed them to functional neurons in adult mouse brains, restoring synaptic connectivity and partial motor function in models, with efficiencies around 5-15% by 4-6 weeks post-injection. Chemical approaches, avoiding genetic , have emerged as alternatives; for instance, small-molecule cocktails applied systemically or locally induced astrocyte-to-neuron conversion in the , yielding electrophysiologically mature neurons by 2-4 weeks without risks. Partial reprogramming, using transient OSK (omitting c-Myc) expression via doxycycline-inducible systems, has shown promise for rejuvenation, extending median lifespan by 30% in mouse models and reversing epigenetic age markers in multiple tissues without tumorigenesis. Applications span , particularly in non-regenerative organs. In the heart, GMT-mediated reprogramming post-injury enhanced vascularization and contractility in mice, with human trials exploring similar vectors as of 2023, though scaled remains a barrier. For repair, Sox2-driven conversion of astrocytes to neuroblasts in injured adult mice generated doublecortin-positive cells by 7 days, contributing to modest functional recovery in hindlimb locomotion assays. Partial protocols have ameliorated age-related declines, such as improved pancreatic and regrowth in naturally aged mice subjected to cyclic OSK for 2 months annually. These strategies leverage the body's native microenvironment to promote integration, potentially outperforming transplanted cells in avoiding immune rejection and vascular mismatches. Challenges include persistently low reprogramming efficiencies (typically 1-20%, varying by tissue and factors), attributed to epigenetic barriers and hostile microenvironments like or , which suppress factor activity.30313-5.pdf) Viral delivery risks and , while chemical methods face issues and off-target effects, as seen in unintended glial proliferation. Tumorigenesis arises from pluripotency factors like c-Myc, prompting safer partial regimens, though long-term heterogeneity—where reprogrammed cells exhibit immature phenotypes or revert—persists . precision remains critical, with non-invasive options like nanoparticles under investigation to target specific regions without . Ongoing refinements, such as combining factors with epigenetic modifiers, aim to boost yields, but clinical lags due to these variabilities and the need for human-relevant models.

Partial vs. Full Reprogramming

Full reprogramming entails the complete of cells into induced pluripotent cells (iPSCs) through sustained expression of the Yamanaka factors—Oct4, , , and c-Myc (OSKM)—resulting in erasure of epigenetic memory, activation of pluripotency networks, and potential for formation upon transplantation. This , first demonstrated in fibroblasts in 2006 and cells in 2007, enables broad differentiation potential but carries substantial risks of genomic instability and oncogenesis due to full dedifferentiation and of pluripotent cells. In contrast, partial reprogramming applies transient or cyclic exposure to subsets of these factors (often OSK, excluding oncogenic c-Myc) or chemical mimics, aiming to reset age-associated epigenetic marks—such as patterns—while preserving cellular identity and function, thereby avoiding pluripotency and associated tumorigenic liabilities. Mechanistically, full reprogramming drives mesenchymal-to-epithelial transition, metabolic shifts to , and global , often requiring 2–4 weeks for iPSC colony emergence with efficiencies below 1% in standard protocols. Partial approaches, however, induce shorter bursts of factor expression (e.g., 2–10 days), triggering selective demethylation at aging-related loci without widespread pluripotency gene , as evidenced by multi-omics showing intermediate epigenetic states between and pluripotent cells. This distinction yields divergent outcomes: fully reprogrammed iPSCs exhibit epigenetic ages akin to embryonic stem cells but demand redifferentiation for therapeutic use, whereas partial methods demonstrably reduce biological age metrics—like Horvath clock estimates—by 20–50% in fibroblasts and extend proliferative capacity without loss of lineage commitment. Empirical evidence underscores partial reprogramming's advantages for . In a 2016 study, transient OSKM expression in progeroid mice improved tissue repair and lifespan without induction, contrasting full reprogramming's pluripotency-driven risks. Subsequent work in 2023–2024 confirmed OSK-mediated partial reprogramming ameliorates hallmarks in human cells, enhances mitochondrial function, and delays aging phenotypes , with cyclic protocols yielding up to 30% lifespan extension in nematodes. Full reprogramming, while foundational for , remains constrained by incomplete epigenetic erasure in ~10–20% of iPSC lines and persistent somatic memory, necessitating partial strategies for safer, identity-preserving applications in aging and disease. Chemical partial reprogramming, emerging in 2025 protocols, further mitigates genetic integration risks by using small molecules to mimic OSK effects, achieving similar in aged human fibroblasts.

Challenges and Criticisms

Efficiency and Variability

Reprogramming processes across experimental methods exhibit notoriously low efficiencies, often below 1%, posing significant barriers to scalability and clinical translation. In defined factor induction using the Yamanaka factors (Oct4, , , and c-Myc), initial retroviral transduction of mouse fibroblasts yielded reprogramming efficiencies of approximately 0.01-0.1%, with human cells showing similar or lower rates due to additional barriers like p53-mediated senescence.00111-1) (SCNT) fares comparably poorly, with cloning efficiencies typically ranging from 1-5% in mammalian models, attributed to incomplete epigenetic remodeling and oocyte-imposed barriers. Cell fusion techniques achieve modestly higher rates than lentiviral iPSC methods (e.g., >0.00025%), but still remain inefficient overall, often requiring hybrid selection that introduces further artifacts. Variability in reprogramming outcomes manifests at multiple levels, including activation of pluripotency networks and heterogeneous epigenetic landscapes among resulting cells. Single-cell analyses reveal substantial cell-to-cell differences in transcriptional trajectories during early iPSC , masked in bulk populations, leading to inconsistent colony formation and potency. Epigenetic predisposition, such as preexisting patterns, further exacerbates this, with somatic cells displaying variable susceptibility based on origin, age, and proliferative state—faster-proliferating cells reprogram more readily, while aged or quiescent ones resist due to entrenched . In SCNT, donor and nuclear envelope breakdown timing contribute to erratic blastocyst development rates, often resulting in aberrant imprinting or X-chromosome reactivation. Proteomic and assays of iPSC derivatives highlight batch-to-batch inconsistencies, with protocols yielding variable ratios and expression profiles that undermine . These inefficiencies and variabilities stem from multifaceted barriers, including transcriptional , DNA damage responses, and incomplete demethylation at CpG sites, which collectively limit the fraction of input cells achieving stable pluripotency. Efforts to mitigate them, such as small-molecule enhancers or miRNA supplementation, have boosted efficiencies up to 100-fold in optimized settings but fail to eliminate underlying stochasticity, as evidenced by persistent heterogeneity in clinical-grade lines.00111-1) Across methods, donor-specific factors like genetic background amplify outcome disparities, with fibroblasts from diverse sources showing up to 10-fold efficiency swings, complicating standardization. Such challenges underscore the need for mechanistic insights into rate-limiting steps, like TET-mediated demethylation, to achieve deterministic reprogramming.

Tumorigenesis Risks

The introduction of reprogramming factors, particularly c-Myc and among the Yamanaka factors (Oct4, , , c-Myc), carries inherent tumorigenic potential due to their roles in promoting and oncogenesis. c-Myc, a proto-oncogene, drives uncontrolled and is implicated in up to 70% of human cancers, with its transient overexpression during iPSC generation linked to increased mutation rates and tumor initiation in mouse models. Klf4 similarly exhibits oncogenic activity in certain contexts, such as enhancing tumor growth in breast and gastrointestinal cancers when dysregulated. Reprogramming induces genomic instability, including copy number variations (CNVs), single variants (SNVs), and chromosomal aberrations, which persist in iPSCs and their derivatives. Studies report CNVs in 12-46% of iPSC lines, often at cancer-associated loci like 17q21 (involving tumor suppressors), arising from replication stress and DNA damage response suppression during the mesenchymal-to-epithelial transition phase. The p53-PUMA axis, a key guardian against genomic damage, is downregulated to facilitate reprogramming efficiency, but this elevates risk, as evidenced by higher incidence in p53-deficient reprogrammed cells. Undifferentiated or partially reprogrammed iPSCs form teratomas—benign tumors with multi-lineage —upon transplantation, a standard confirming pluripotency but highlighting clinical peril. , residual pluripotent cells in therapeutic grafts, even at low frequencies (<0.001%), can proliferate into malignant teratocarcinomas, as demonstrated in models where iPSC-derived cells caused tumors in 20-30% of recipients. Heterogeneity in reprogrammed populations exacerbates this, with subpopulations retaining epigenetic or oncogenic signatures prone to neoplastic transformation under stress. Efforts to quantify long-term risk include tracking iPSC-derived cardiomyocytes, where 1-5% exhibit arrhythmogenic potential linked to latent tumorigenicity, though human trials remain limited by these concerns as of 2023. Non-integrating methods (e.g., mRNA delivery) reduce but do not eliminate factor-induced instability or propensity. Overall, while mitigation strategies like suicide genes or purification exist, tumorigenesis remains a core barrier to safe iPSC translation, underscored by regulatory scrutiny from bodies like the FDA emphasizing preclinical tumor assays.

Incomplete Reprogramming and Heterogeneity

Incomplete reprogramming during the generation of induced pluripotent stem cells (iPSCs) occurs when epigenetic marks, such as patterns and modifications, are not fully erased despite expression of reprogramming factors like the Yamanaka factors (Oct4, , , and c-Myc). This retention of "epigenetic memory" biases iPSCs toward their tissue of origin, manifesting as incomplete resetting of profiles and states. For instance, iPSCs derived from fibroblasts or cells show hypermethylation at somatic-specific promoters, leading to inefficient activation of pluripotency networks and persistent lineage-specific gene signatures. Such memory can be partially mitigated through serial reprogramming or targeted demethylation treatments, but it persists in many lines, contributing to functional variability. Heterogeneity in reprogramming arises from activation of pluripotency genes across cells, resulting in a mixed population where only a subset achieves full pluripotency while others remain in intermediate states. Single-cell sequencing during Yamanaka reveals asynchronous waves of transcriptional changes, with early responders progressing faster but late-phase cells often stalling due to epigenetic barriers like Polycomb-mediated repression. This variability is exacerbated by donor ; for example, iPSCs from differentiated lineages exhibit greater heterogeneity in timing compared to embryonic stem cells, with up to 20-30% of domains retaining somatic-like profiles. Studies of multiple iPSC clones confirm line-to-line differences in fidelity, influenced by factors such as reprogramming duration and vector integration, leading to non-uniform potential. The consequences of incomplete reprogramming and heterogeneity include biased —e.g., pancreatic iPSCs preferentially form insulin-producing s—and increased of aberrant lineages or genomic in downstream applications. In clinical contexts, this manifests as clone-specific variability, necessitating extensive screening; for instance, only select clones from heterogeneous pools show comparable potency to embryonic stem s in teratoma formation assays. Efforts to reduce heterogeneity, such as using naive pluripotency media or auxiliary factors like H1FOO, have improved uniformity but do not eliminate residual in all cases. Overall, these issues highlight technical limitations in achieving a true ground-state reset, with epigenetic profiling recommended for validating iPSC quality.

Ethical and Societal Considerations

Advantages Over Embryonic Stem Cells

Induced pluripotent stem cells (iPSCs), generated through reprogramming of somatic cells via defined factors such as Oct4, Sox2, Klf4, and c-Myc, offer ethical advantages over embryonic stem cells (ESCs) by circumventing the need to derive cells from human embryos, thereby avoiding the destruction of embryonic tissue inherent in ESC isolation. This approach aligns with concerns raised since the 1990s regarding the moral status of embryos, enabling research and therapeutic applications without the associated controversies that have limited ESC funding and policy support in various jurisdictions. A primary immunological benefit of iPSCs is their potential for autologous use, where cells are reprogrammed from a patient's own tissues, such as fibroblasts, minimizing risks of immune rejection that plague allogeneic ESC-derived therapies requiring immunosuppressive drugs. This patient-specific derivation facilitates , including disease modeling and drug screening tailored to individual genetic backgrounds, as demonstrated in studies generating iPSCs from patients with conditions like since the technique's establishment in 2006 by Yamanaka and colleagues. Practically, iPSCs provide greater accessibility and scalability, as they can be produced from abundant adult sources without reliance on limited supplies or the technical challenges of oocyte donation and used in some alternatives. iPSCs maintain pluripotency comparable to ESCs, with indefinite and into all three layers, but their generation avoids the ethical and logistical barriers of procurement, accelerating timelines—evidenced by over 1,000 iPSC lines established globally by 2012 for diverse applications. This has enabled broader adoption in , though full clinical equivalence to ESCs remains under validation through comparative genomic and functional assays.

Hype, Funding, and Unfulfilled Promises

The discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka's team in 2006 generated intense excitement, positioning reprogramming as a transformative technology capable of generating patient-matched cells for regenerative therapies while sidestepping ethical issues tied to derivation. Researchers and media outlets heralded iPSCs as a "" for cells, forecasting rapid cures for conditions like , injuries, and through personalized tissue regeneration. This optimism fueled visions of democratizing applications, with early demonstrations of reprogramming fibroblasts using four transcription factors (Oct4, , , and c-Myc) suggesting broad applicability across species and cell types. The enthusiasm spurred massive funding commitments. In 2004, California voters approved Proposition 71, allocating $3 billion over 10 years to the California Institute for Regenerative Medicine (CIRM) for initiatives, which expanded post-iPSC discovery to include reprogramming research; a 2020 ballot measure (Proposition 14) added $5.5 billion in bonds, with $1.5 billion earmarked for neurological applications. Federally, the U.S. (NIH) has invested hundreds of millions annually in research, totaling over $2 billion from fiscal years 2013 to 2025, much of it supporting iPSC-related projects amid global private sector inflows exceeding $10 billion cumulatively by the mid-2010s. These resources enabled thousands of publications and preclinical studies but were often justified by projections of imminent clinical breakthroughs. Despite the investments, reprogramming's clinical impact remains limited as of 2025, with unfulfilled promises stemming from persistent technical barriers rather than mere incremental progress. While iPSCs have excelled as research tools for disease modeling and drug screening—evident in adoption for humanized assays—therapeutic translation has lagged, with no fully approved iPSC-derived products for routine use due to issues like incomplete epigenetic resetting, genetic instability, and manufacturing scalability. Early trials, such as those for using iPSC-derived retinal cells initiated around 2014, demonstrated feasibility but faced setbacks including tumor risks and variable efficacy, underscoring how initial hype overlooked the causal complexities of safe, reproducible reprogramming in humans. A 2025 review of pluripotent trials notes only modest advancements in niche areas like vision restoration, attributing delays to regulatory hurdles and empirical failures in potency and , prompting critiques that funding has disproportionately supported exploratory work over validated pipelines. This gap highlights a pattern where optimistic projections, amplified by academic and media narratives, have not materialized into widespread societal benefits, raising questions about resource allocation in fields prone to overstatement.

Transgenerational and Environmental Concerns

Reprogramming of cells to induced pluripotent stem cells (iPSCs) entails global epigenetic reconfiguration, including and histone modifications, which theoretically could leave residual marks transmissible through the if iPSCs or derivatives are used in reproductive contexts. However, mammalian development naturally imposes two waves of epigenetic reprogramming to erase imprints, minimizing inheritance of acquired marks, as evidenced by single-cell analyses showing near-complete demethylation in human cells.00629-7) Despite this, iPSCs exhibit persistent epigenetic from donor cell types, with source-specific patterns retained at loci influencing propensity, such as bivalent promoters in fibroblasts-derived iPSCs favoring mesodermal lineages. This diminishes with extended passaging or serial reprogramming but raises hypothetical risks for heritable biases if iPSC-derived gametes transmit incomplete erasure, though in mammals remains absent due to ethical constraints on human editing. itself is contentious in vertebrates, with most reported cases limited to paramutation-like effects in or short-term phenomena in , and human studies failing to demonstrate stable transmission beyond parental effects confounded by cultural or exposures. Environmental factors modulate reprogramming outcomes by altering accessibility and factor binding, with stressors like or toxins influencing Yamanaka factor (OSKM) efficacy and epigenetic fidelity. For instance, exposure to endocrine disruptors during iPSC derivation can induce heritable changes in offspring via sperm miRNA-mediated mechanisms in model systems, though direct links to reprogramming protocols are unestablished. In production contexts, iPSC manufacturing demands high-resource bioreactors and serum-free media, potentially amplifying ecological footprints through energy-intensive sterile culturing and waste from viral vectors or chemical inducers, yet quantitative assessments indicate lower impacts than lines due to non-embryonic sourcing. Conversely, iPSC scalability for or conservation biobanking offers mitigation of livestock-related emissions, with projections estimating up to 99% reduction in land use for protein production, underscoring net environmental benefits over concerns if optimized. Rigorous assessment of these interactions remains essential, as microenvironmental heterogeneity in aggregates can perpetuate epigenetic variability, complicating therapeutic predictability.

Recent Developments

mRNA and Non-Integrating Approaches

Non-integrating approaches to cellular reprogramming deliver reprogramming factors without incorporating foreign DNA into the host genome, thereby avoiding and enabling footprint-free induced pluripotent stem cells (iPSCs). These methods include synthetic mRNA , protein , episomal vectors, and non-integrating RNA viruses such as Sendai virus, which express Yamanaka factors (Oct4, , , and c-Myc) transiently before degradation. Compared to integrating lentiviral or retroviral systems, non-integrating techniques yield lower but safer reprogramming rates, with efficiencies typically ranging from 0.001% to 0.1% depending on and protocol, though they reduce tumorigenic risks associated with persistent expression. mRNA-based reprogramming emerged as a prominent non-integrating in 2010, when Warren et al. demonstrated highly efficient of fibroblasts to iPSCs using synthetic modified mRNAs encoding the Yamanaka factors, achieving up to 4.4-fold higher efficiency than comparable DNA-based methods through repeated transfections over 14-16 days.00434-0) Chemical modifications, such as substitution and 5-methylcytidine capping, suppress Toll-like receptor-mediated immune responses that degrade unmodified mRNA, allowing sustained protein expression without viral vectors. This approach generates transgene-free iPSCs verified by pluripotency markers like Tra-1-60 and SSEA-4, with successful into lineages such as neurons and cardiomyocytes.00434-0) Among non-integrating methods, mRNA transfection consistently outperforms alternatives in speed and yield; for instance, a 2014 comparative study of human fibroblasts found mRNA yielding colonies in 14 days at 0.05-0.2% efficiency, versus 25-30 days for Sendai virus or episomes at lower rates, attributed to mRNA's rapid translation and short half-life (hours to days). Sendai virus, an RNA paramyxovirus, provides prolonged expression (up to 10 passages) without integration but requires BSL-2 handling and may leave residual viral RNA detectable by PCR. Episomal vectors, such as oriP/EBNA1-based plasmids, persist extrachromosomally but dilute unevenly during division, often necessitating selection markers. Protein-based delivery of recombinant Yamanaka factors avoids nucleic acids entirely but suffers from poor stability and membrane permeability, limiting efficiency to below 0.01%. Recent advancements leverage mRNA for partial reprogramming, aiming to reverse aging phenotypes without inducing pluripotency. In a 2020 study, transient mRNA delivery of OSKM factors to adult human fibroblasts restored youthful epigenetic clocks, enhanced nucleocytoplasmic compartmentalization, and reduced DNA damage accumulation after four rounds of transfection, without tumorigenic transformation. By 2025, optimized xeno-free mRNA protocols have enabled scalable iPSC production for regenerative medicine, with efficiencies approaching 1% in optimized fibroblast lines, though challenges persist in primary cells like blood progenitors due to transfection toxicity. These methods support applications in longevity research, where cyclic low-dose mRNA dosing mitigates senescence in progeria models, highlighting their potential for transient epigenetic modulation over full reprogramming.

Clinical Trials and Regenerative Medicine

Clinical trials involving induced pluripotent stem cell (iPSC)-derived therapies have primarily focused on phase I and II studies assessing safety and preliminary efficacy in regenerative applications, with over 115 trials approved worldwide as of December 2024 testing 83 human pluripotent stem cell products, predominantly targeting ophthalmological, neurological, and cardiovascular conditions.00445-4) These trials leverage reprogramming to generate patient-specific or allogeneic cells for tissue repair, bypassing ethical issues associated with embryonic sources while aiming to restore function in degenerative diseases. Early data indicate tolerable safety profiles in most cases, though long-term efficacy remains under evaluation due to the nascent stage of many protocols.00445-4) In , iPSC-derived epithelial (RPE) cells have shown promise for age-related (AMD); Japan's first-in-human autologous , initiated in 2014 by , transplanted iPSC-RPE sheets into patients, reporting no serious adverse events and modest visual improvements in some participants over two years, though the program shifted to allogeneic approaches due to manufacturing scalability issues. Subsequent allogeneic , such as those by Lineage Cell Therapeutics (NCT05176761), have advanced to phase I/II, demonstrating graft survival via imaging without tumor formation in initial cohorts as of 2024. For , a phase I (NCT05635409) using allogeneic iPSC-derived neurons reported safe implantation in the with no graft-derived tumors at one-year follow-up, though motor symptom efficacy requires further validation in larger studies.00445-4) Cardiovascular applications include iPSC-cardiomyocyte patches for ischemic ; a Japanese phase I trial (2018-2023) involving intramyocardial injection of allogeneic sheets in 12 patients yielded improved left ventricular function ( increase of ~10%) and reduced infarct size on MRI at one year, with no arrhythmias or malignancies observed.00053-2/fulltext) In spinal cord injury, iPSC-derived mesenchymal stromal cells in a phase I/IIa trial showed 60% participant survival at two years post-transplantation, outperforming historical controls, alongside modest sensory improvements in subsets, though motor gains were inconsistent. Diabetes trials, such as ' phase I/II (NCT04786262) using allogeneic iPSC-islet cells, reported insulin independence in one patient after 90 days as of updates, highlighting potential for beta-cell replacement but noting needs. Despite progress, challenges persist: tumorigenesis risks from residual pluripotency, immune rejection in allogeneic settings, and variable engraftment efficiency have led to cautious trial designs, with no phase III approvals for iPSC therapies as of 2025. Regulatory bodies like Japan's PMDA and the FDA emphasize off-the-shelf allogeneic models for scalability, yet potency assays and long-term monitoring remain hurdles, as evidenced by halted autologous programs due to high costs exceeding $1 million per patient. Ongoing trials prioritize non-integrating reprogramming to minimize genomic risks, supporting broader regenerative potential.

Rejuvenation and Longevity Research

Cellular reprogramming techniques, particularly partial reprogramming using subsets of the Yamanaka factors (OCT4, , , or OSK), aim to reverse epigenetic aging signatures while preserving cellular identity, thereby targeting such as epigenetic drift and loss of information. This approach resets patterns and to a more youthful state, as demonstrated in progeroid mice where transient OSKM expression improved tissue function and extended lifespan. Unlike full reprogramming to induced pluripotent stem cells, partial methods avoid tumorigenic risks by limiting factor exposure duration, often via doxycycline-inducible systems. In vivo studies in mice have shown systemic partial reprogramming can ameliorate multiple age-related phenotypes. For instance, inducible OSK delivered via adeno-associated viruses in 124-week-old male mice extended median lifespan by approximately 10% and reduced age-related pathologies in tissues like and . Similarly, epigenetic restoration experiments reversed aging signs, including improved vision in glaucoma-damaged optic nerves and reduced , by repairing information loss rather than DNA mutations. These effects correlate with epigenetic clock reversal, where biological age markers like Horvath's clock decrease post-reprogramming. Chemical-based reprogramming offers a non-genetic , using small-molecule cocktails to mimic Yamanaka effects. A 2023 Harvard study identified compounds that restored youthful transcriptomes and epigenetic ages in senescent human fibroblasts and cells, delaying without vectors. More recent work in 2025 demonstrated that defined chemical mixtures rejuvenated aged human cells, extended their replicative lifespan, and reversed mesenchymal drift—a prevalent aging feature linked to —prior to pluripotency induction.00853-0) Progress toward human application remains preclinical, with no completed trials as of 2025, though companies like Rejuvenate Bio and Life Biosciences are advancing OSK-based therapies for age-related diseases. Early-phase trials for partial reprogramming in and are in preparation, building on vision restoration data from 2020. Challenges include optimizing dosage to maximize while minimizing off-target effects, as over-expression risks incomplete epigenetic erasure or heterogeneity. Ongoing research emphasizes multi-omics validation to confirm causal links between reprogramming and extension.

References

  1. [1]
    Cellular Reprogramming - PMC - NIH
    It can rejuvenate somatic cells by erasing the epigenetic memories and reconstructing a new pluripotent order.
  2. [2]
    What is Cell Reprogramming and What Does It Mean? - BioInformant
    Rating 4.9 (16) Dec 13, 2024 · Cell reprogramming is the act of reverting mature, specialised cells into induced pluripotent stem cells, also known as iPS cells.
  3. [3]
    Induction of pluripotent stem cells from mouse embryonic and adult ...
    Differentiated cells can be reprogrammed to an embryonic-like state by transfer of nuclear contents into oocytes or by fusion with embryonic stem (ES) cells ...
  4. [4]
    Epigenetics of Cellular Reprogramming - PMC - PubMed Central - NIH
    Epigenetic reprogramming involves changes in chromatin structure and gene expression, driven by epigenetic modulators, to change cell identity.
  5. [5]
    Induced pluripotent stem cells (iPSCs): molecular mechanisms of ...
    Apr 26, 2024 · Eventually, Takahashi and Yamanaka identified a combination of 4 reprogramming factors—Oct4, Sox2, Klf4, and Myc—that was sufficient to ...
  6. [6]
    Cellular reprogramming and epigenetic rejuvenation
    Sep 6, 2021 · For a cellular reprogramming-based intervention to be considered rejuvenative (turning an old cell into a younger cell), we need to uncouple its ...
  7. [7]
    Mechanisms, pathways and strategies for rejuvenation through ...
    Dec 15, 2023 · Here we discuss the current state of epigenetic rejuvenation strategies via partial reprogramming in both mouse and human models.
  8. [8]
    DNA Methylation Reprogramming during Mammalian Development
    Mar 29, 2019 · Epigenetic reprogramming in early embryos involves erasure of most methylation marks inherited from the gametes (exceptions include methylation ...
  9. [9]
    Rebooting the Epigenomes during Mammalian Early Embryogenesis
    Here, we review the recent progress in the understanding of epigenetic reprogramming in early embryos and how it contributes to the mammalian early development.
  10. [10]
    Active and Passive Demethylation of Male and Female Pronuclear ...
    Oct 2, 2014 · Here we show that both maternal and paternal genomes undergo widespread active and passive demethylation in zygotes before the first mitotic division.
  11. [11]
    DNA methylation dynamics during epigenetic reprogramming of ...
    Apr 22, 2019 · In mammals, global genomic DNA methylation levels of the zygote decrease after fertilization and reach the lowest level at blastula stage [24–27] ...
  12. [12]
    Active demethylation of the paternal genome in the mouse zygote
    The TKZ751 and myl-C genes were highly methylated in both mature oocytes and sperm, whereas Igf2 and α-actin were methylated in mature sperm but not in oocytes ...Missing: fertilization | Show results with:fertilization
  13. [13]
    GSE is a maternal factor involved in active DNA demethylation in ...
    The dynamic epigenetic changes during early embryo development primarily involve DNA methylation and demethylation.
  14. [14]
    Genome-coverage single-cell histone modifications for embryo ...
    Feb 26, 2025 · After fertilization, cells undergo considerable epigenetic reprogramming for zygotic genome activation (ZGA) and lineage specification to ...
  15. [15]
    Review Epigenetic regulation of early human embryo development
    Dec 7, 2023 · Recent studies have now produced the first epigenetic maps of early human embryogenesis, stimulating new ideas about epigenetic reprogramming, cell fate ...
  16. [16]
    DNA methylation and memory formation | Nature Neuroscience
    Oct 26, 2010 · Recent evidence indicates that DNA methylation may serve as a contributing mechanism in memory formation and storage.
  17. [17]
    DNA methylation changes in plasticity genes accompany ... - Nature
    Dec 14, 2015 · We found that histone modifications predominantly changed during memory acquisition and correlated surprisingly little with changes in gene ...
  18. [18]
    Dnmt3a1 regulates hippocampus-dependent memory via ... - Nature
    Mar 18, 2024 · Specifically, DNA methylation is necessary for the formation of long-term memory in multiple brain regions including the hippocampus. Despite ...
  19. [19]
    Neuronal ensemble-specific DNA methylation strengthens engram ...
    Jan 31, 2020 · DNA methylation changes in plasticity genes accompany the formation and maintenance of memory. Nat. Neurosci. 19, 102–110 (2016). Article ...
  20. [20]
    Epigenetics in Learning and Memory - PubMed
    The data support a model in which epigenetic modifications in promoters and enhancers facilitate the priming and activation of these regulatory regions, hence ...
  21. [21]
    Epigenetic Regulation of Learning and Memory by Drosophila ...
    Our study identifies Drosophila EHMT as a key regulator of cognition that orchestrates an epigenetic program featuring classic learning and memory genes.
  22. [22]
    Researchers rejuvenate brain neurons through cellular ...
    A research study led by the University of Barcelona describes how brain neurons in mice can be rejuvenated through a controlled cellular reprogramming cycle.
  23. [23]
    Epigenetic regulation of brain development, plasticity, and response ...
    Aug 6, 2025 · Epigenetic mechanisms, including DNA methylation and chromatin modifications, can confer both stability and dynamic plasticity for developing ...
  24. [24]
    Epigenetic rejuvenation by partial reprogramming - PubMed
    Mar 4, 2023 · Recent studies indicate that partial reprogramming by limited exposure to reprogramming factors can reset epigenetic ageing clocks while maintaining cellular ...
  25. [25]
    Reprogramming to recover youthful epigenetic information ... - Nature
    Dec 2, 2020 · In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice ... David A. Sinclair. View author ...
  26. [26]
    Gene Therapy-Mediated Partial Reprogramming Extends Lifespan ...
    Recent studies have demonstrated that partial reprogramming using the Yamanaka factors (or a subset; OCT4, SOX2, and KLF4; OSK) can reverse age-related changes ...
  27. [27]
    Chemically induced reprogramming to reverse cellular aging
    Jul 12, 2023 · We identify six chemical cocktails, which, in less than a week and without compromising cellular identity, restore a youthful genome-wide transcript profile ...
  28. [28]
    Chemically induced reprogramming to reverse cellular aging
    The ability of the Yamanaka factors to erase cellular identity raised a key ... rejuvenation mechanisms are shared between the two modes of partial reprogramming.
  29. [29]
    A single factor for safer cellular rejuvenation - bioRxiv
    Jun 6, 2025 · Our platform identified SB000, the first single gene intervention to rejuvenate cells from multiple germ layers with efficacy rivalling the Yamanaka factors.
  30. [30]
    Partial cellular reprogramming: A deep dive into an emerging ...
    Partial cellular reprogramming is able to improve or restore multiple age‐related phenotypes including lifespan, healthspan, epigenetic age, aging hallmarks, ...
  31. [31]
    Epigenetic reprogramming as a key to reverse ageing and increase ...
    Jan 23, 2024 · Notably, partial reprogramming enables the resetting of the ageing clock without erasing cellular identity.
  32. [32]
    Rejuvenate Bio Announces Gene Therapy-Mediated Partial ...
    Feb 28, 2025 · Rejuvenate Bio's research focuses on partial reprogramming using the Yamanaka factors, specifically OCT4, SOX2, and KLF4 (OSK), to reverse age-related changes.
  33. [33]
  34. [34]
    An epigenomic roadmap to induced pluripotency reveals DNA ...
    Dec 10, 2014 · We find that DNA methylation gain during reprogramming occurs gradually, while loss is achieved only at the ESC-like state. Binding sites of ...
  35. [35]
    Epigenetic reprogramming of cell identity: lessons from development ...
    Jul 23, 2021 · Epigenetic mechanisms are known to define cell-type identity and function. Hence, reprogramming of one cell type into another essentially ...
  36. [36]
    Progress in understanding reprogramming to the induced ...
    Reprogramming to pluripotency upon Oct4, Sox2, Klf4, and cMyc overexpression takes at least one to two weeks for the first reprogrammed cells to emerge in the ...
  37. [37]
    Human transcription factors responsive to initial reprogramming ...
    Nov 12, 2020 · The four transcription factors OCT4, SOX2, KLF4, and MYC (OSKM) together can convert human fibroblasts to induced pluripotent stem cells (iPSCs).
  38. [38]
    The Role of DNMT Methyltransferases and TET Dioxygenases in the ...
    Sep 4, 2024 · This review deals with the functional characteristics and biological roles of enzymes participating in DNA methylation and demethylation as key factors in ...
  39. [39]
    The occurrence and development of induced pluripotent stem cells
    The second possible mechanism is a random model (Figure 1) (Yamanaka, 2009). This theory suggests that OSKM can reprogram all mediated somatic cells in a fixed ...
  40. [40]
    Molecular Control of Induced Pluripotency - ScienceDirect.com
    Jun 5, 2014 · Here we review recent advances in epigenetic reprogramming to pluripotency with a focus on the principal molecular regulators.
  41. [41]
  42. [42]
    Transcription factor code in iPSC reprogramming - PMC
    Jul 8, 2021 · The understanding of iPSC reprogramming provides insights into the mechanisms that safeguard somatic cell identity, drive epigenetic ...
  43. [43]
  44. [44]
    Optimal Ratio of Transcription Factors for Somatic Cell ... - NIH
    Significance: The overall gene expression profiles between the high and low efficiency conditions provide novel insights for somatic cell reprogramming.
  45. [45]
    Dissecting the impact of transcription factor dose on cell ... - Nature
    Oct 3, 2025 · Leveraging single-cell resolution, we uncovered how TF dose shapes reprogramming heterogeneity, revealing both dose-dependent and stochastic ...
  46. [46]
    Nuclear Cloning and Direct Reprogramming - ScienceDirect.com
    Dec 7, 2012 · In 1952 Briggs and King performed the first successful nuclear transfer (NT) experiments in the frog Rana pipiens. When nuclei from cleavage ...
  47. [47]
    Somatic Cell Nuclear Transfer Reprogramming: Mechanisms and ...
    Oct 4, 2018 · John Gurdon was the first to demonstrate that animals could be cloned from differentiated frog somatic cells by SCNT (Gurdon, 1962; Figure 1).
  48. [48]
    [PDF] Sir John B. Gurdon - Nobel Lecture: The Egg and the Nucleus
    Design of a somatic cell nuclear transfer experiment using unfertilised eggs as first designed by Briggs and King (1952) for Rana pipiens and as used ...
  49. [49]
    Sir John Gurdon: Godfather of cloning - PMC - NIH
    Apr 21, 2008 · Sir John Gurdon is known as the "Godfather of cloning" for his frog cloning experiments, which showed adult cells are genetically identical to ...
  50. [50]
    Use of somatic cell fusion to reprogram globin genes - ScienceDirect
    Somatic cell fusion@21S omatic cell fusion , pioneered by Henry Harris and Nils Ringertz over three decades ago,1–5has been used to illuminate many ...
  51. [51]
    [PDF] Kinetics of reprogramming in cell fusion hybrids
    Feb 16, 2011 · The reestablishment of new characteristics by cell fusion between two different cell types was suggested as early as in. 1965 (Harris and ...
  52. [52]
  53. [53]
    Reprogramming mediated by stem cell fusion - Wiley Online Library
    Advances in mammalian cloning prove that somatic nuclei can be reprogrammed to a state of totipotency by transfer into oocytes. An alternative approach to ...<|control11|><|separator|>
  54. [54]
    The 2012 Nobel Prize in Physiology or Medicine - Advanced ...
    Shinya Yamanaka for their discovery that mature, differentiated cells can be reprogrammed to a pluripotent stem cell state. This represents a paradigm shift in ...
  55. [55]
    Reprogramming to pluripotency: stepwise resetting of the epigenetic ...
    Feb 15, 2011 · In 2006, the “wall came down” that limited the experimental conversion of differentiated cells into the pluripotent state.
  56. [56]
  57. [57]
    Epigenetic reprogramming and induced pluripotency - PMC - NIH
    In this manuscript, we set recent advances in the induction of pluripotency in somatic cells with transcription factors into historical context and discuss the ...
  58. [58]
    Time-course transcriptome analysis of human cellular ...
    Jan 3, 2018 · Briefly, reprogramming is initiated with mesenchymal-to-epithelial transition (MET), one of the hallmark events of initiation. Next, the ...
  59. [59]
    Phases of reprogramming - ScienceDirect.com
    We will review the key cellular and molecular events associated with the reprogramming process, giving an integrative and conciliatory view of the different ...
  60. [60]
    Induced pluripotency: history, mechanisms, and applications
    The generation of induced pluripotent stem cells (iPSCs) from somatic cells demonstrated that adult mammalian cells can be reprogrammed to a pluripotent state.<|separator|>
  61. [61]
    Unique molecular events during reprogramming of human somatic ...
    Upon dox treatment, morphological changes occurred in hiF-T cells at approximately day 2, and small cell aggregates were observed as early as day 6 (Figure 1A).
  62. [62]
    Single-Cell Expression Analyses during Cellular Reprogramming ...
    Stochastic gene expression early in reprogramming is followed by a late hierarchical phase with Sox2 being the upstream factor in a gene expression hierarchy.
  63. [63]
    Induced Pluripotent Stem Cells: Reprogramming Platforms ... - NIH
    Somatic cell reprogramming is initiated by changes in the transcriptome and chromatin structure of differentiated state into that of a pluripotent-like state.
  64. [64]
    Maturation, not initiation, is the major roadblock during ... - PNAS
    These data demonstrate that maturation, and not initiation, is the limiting step during the direct reprogramming of human fibroblasts toward pluripotency.
  65. [65]
    Reprogramming mechanisms influence the maturation of ... - Nature
    Oct 24, 2018 · Multifactorial approach is needed to unravel the maturation phases of human neurons derived from induced pluripotent stem cells. Article Open ...
  66. [66]
    The long and winding road of reprogramming-induced rejuvenation
    Mar 2, 2024 · Another study has shown the capacity to reprogram human somatic cells to pluripotent stem cells using chemicals. Unlike OSKM-mediated ...<|separator|>
  67. [67]
    Transcriptional and epigenetic mechanisms of cellular ... - NIH
    Enforced ectopic expression of a cocktail of pluripotency-associated genes such as Oct4, Sox2, Klf4 and c-Myc can reprogram somatic cells into induced ...
  68. [68]
    A late transition in somatic cell reprogramming requires regulators ...
    Dec 7, 2012 · We show that the stabilization phase and the acquisition of pluripotency are dependent on the removal of transgene expression late in the maturation phase.
  69. [69]
    Metabolic control of induced pluripotency - Frontiers
    By the beginning of the stabilization phase, the activation of additional pluripotent genes, such as Sox2, Utf1, Lin28a, Esrrb, Klf5, Dppa2, and Dppa4, as well ...
  70. [70]
    Mechanisms and models of somatic cell reprogramming - PMC
    This study suggests that the transition to the stabilization phase upon transgene removal is dependent on regulatory pathways distinct from those controlling ...
  71. [71]
    Safe and stable generation of induced pluripotent stem cells using ...
    At the stabilization phase, bona fide iPSC colonies transition to a state of transgene independence. ... Overall, iPSC reprogramming remains inefficient as a ...
  72. [72]
    (PDF) Phases of reprogramming - ResearchGate
    Aug 9, 2025 · ... The last stage of iPSC reprogramming is the stabilization phase, characterized by Tg silencing and capacity of the cells to maintain ...Abstract And Figures · References (80) · Recommended Publications
  73. [73]
    Somatic Cell Nuclear Transfer Reprogramming - NIH
    Oct 4, 2019 · Somatic cell nuclear transfer (SCNT) is an artificial method to confer totipotency, involving injecting a donor cell nucleus into an enucleated ...
  74. [74]
    History of Cell Reprogramming - News-Medical
    In 1962: John Gurgeon is the first person to practically define the cell reprogramming technique in 1962. His research on cloning of frogs through nuclear ...
  75. [75]
    Recent advancements in cloning by somatic cell nuclear transfer
    Jan 5, 2013 · Here we review recent progress in somatic cell cloning, with a special emphasis on epigenetic studies using the laboratory mouse as a model.
  76. [76]
    Epigenetic Reprogramming During Somatic Cell Nuclear Transfer
    Mar 17, 2020 · In this review, we present our understanding of SCNT-mediated nuclear reprogramming ... reprogramming of somatic cell nuclear transfer embryos in ...Abstract · Introduction · Incomplete Epigenetic... · Strategies for Enhancing the...
  77. [77]
    Lessons Learned from Somatic Cell Nuclear Transfer - PMC
    Somatic cell nuclear transfer is used primarily for generating cells and tissues that are immunocompatible with the somatic cell donor, a concept known as ...
  78. [78]
    Efficient Somatic Cell Nuclear Transfer by Overcoming Both Pre ...
    Jul 8, 2025 · Mammalian cloning through somatic cell nuclear transfer (SCNT) involves reprogramming terminally differentiated cells into totipotent ...
  79. [79]
    Cell fusion-induced reprogramming - PubMed
    Genomic reprogramming can be accomplished by five different types of methods: nuclear transfer, cell fusion, in vitro culture, introduction of egg extract, ...
  80. [80]
    Nuclear reprogramming in heterokaryons is rapid, extensive, and ...
    Jan 13, 2009 · Helen M. Blau. Corresponding Author. E-mail address: hblau ... reprogramming of cells following fusion has not been assessed to date.
  81. [81]
    Reprogramming to a muscle fate by fusion recapitulates differentiation
    Fusion of mammalian cells to form stable, non-dividing heterokaryons results in nuclear reprogramming without the exchange of genetic material.
  82. [82]
    A Cell Electrofusion Chip for Somatic Cells Reprogramming
    Cell fusion is a potent approach to explore the mechanisms of somatic cells reprogramming. However, previous fusion methods, such as polyethylene glycol ...
  83. [83]
    Cell Fusion-Induced Reprogramming - Springer Nature Experiments
    Genomic reprogramming can be accomplished by five different types of methods: nuclear transfer, cell fusion, in vitro culture, introduction of egg extract, and ...
  84. [84]
    DNA Synthesis Is Required for Reprogramming Mediated by Stem ...
    Embryonic stem cells (ESCs) can instruct the conversion of differentiated cells toward pluripotency following cell-to-cell fusion by a mechanism that is ...Missing: techniques | Show results with:techniques
  85. [85]
    Reprogramming lineage identity through cell–cell fusion
    Jun 2, 2021 · Cell–cell fusion provides an ideal model system for identifying the earliest events in reprogramming.
  86. [86]
    Nuclear reprogramming to a pluripotent state by three approaches
    Jun 10, 2010 · This has been shown by three distinct experimental approaches to nuclear reprogramming: nuclear transfer, cell fusion and transcription-factor transduction.
  87. [87]
    Stem-cell fusion: A twist of fate - Nature
    Oct 3, 2002 · Stem-cell fusion: A twist of fate. Helen M. Blau. Nature ... Cell fusion has long been known to achieve effective reprogramming of cells.
  88. [88]
    Reprogramming mediated by stem cell fusion - PMC
    Potential fusion partners with reprogramming activities include embryonic stem cells, embryonic germ cells, embryonal carcinoma cells, and even differentiated ...Missing: methods | Show results with:methods
  89. [89]
    (PDF) Yamanaka, S. & Blau, H.M. Nuclear reprogramming to a ...
    Aug 9, 2025 · An adult cell can therefore be reprogrammed, altering its pattern of gene expression, and hence its fate, to that typical of another cell type.<|separator|>
  90. [90]
    Induced Pluripotent Stem Cell Experiments by Kazutoshi Takahashi ...
    Jun 1, 2015 · In 2006, Kazutoshi Takahashi and Shinya Yamanaka reprogrammed mice fibroblast cells, which can produce only other fibroblast cells, to become ...
  91. [91]
    Induction of pluripotent stem cells by defined factors is greatly ... - NIH
    We report that DNA methyltransferase and histone deacetylase (HDAC) inhibitors improve reprogramming efficiency.
  92. [92]
    Induction of pluripotent stem cells from adult human fibroblasts by ...
    Nov 30, 2007 · Here, we demonstrate the generation of iPS cells from adult human dermal fibroblasts with the same four factors: Oct3/4, Sox2, Klf4, and c-Myc.
  93. [93]
    Progress in the Reprogramming of Somatic Cells
    Feb 1, 2013 · Mechanisms of reprogramming to pluripotency induced by exogenous transcription factors (TFs) and hypothetical mechanisms of small molecule ...
  94. [94]
    Cell reprogramming: methods, mechanisms and applications
    Mar 27, 2025 · Cell reprogramming is a complex and ordered process that involves the coordination of transcriptional, epigenetic, translational and metabolic changes.<|separator|>
  95. [95]
    Rapid and efficient reprogramming of somatic cells to induced ...
    Somatic cells can be reprogrammed to induced pluripotent stem cells (iPSCs) by expressing four transcription factors: Oct4, Sox2, Klf4, and c-Myc.Results · Mouse Ipscs Produced By... · Human Es And Ipsc Culture
  96. [96]
    Induced Pluripotent Stem (iPS) Cell Culture Methods and ... - NIH
    iPS cells can be cultured in feeder-layer or feeder-free conditions. Here, we describe the iPS cell culture methods and induction differentiation of these ...
  97. [97]
    Guidelines and Techniques for the Generation of Induced ...
    Dec 4, 2008 · We compare the currently reported protocols, identify essential steps common to these methods, and suggest minimal criteria for defining fully reprogrammed ...Protocol Review · Main Text · Choice Of Reprogramming...
  98. [98]
    Complete suspension culture of human induced pluripotent stem ...
    Nov 12, 2024 · This comprehensive and compelling study presents a robust, cost-effective method for expanding pluripotent stem cells.
  99. [99]
    Study reports first ever in vivo production of stem cells - ecancer
    Sep 11, 2013 · Study reports first ever in vivo production of stem cells ... Reprogramming in vivo produces teratomas and iPS cells with totipotency features.Missing: date | Show results with:date
  100. [100]
    Direct Cardiac Reprogramming | Circulation Research
    Sep 13, 2013 · In vivo cardiac cellular reprogramming efficacy is enhanced by angiogenic preconditioning of the infarcted myocardium with vascular ...
  101. [101]
    In vivo chemical reprogramming of astrocytes into neurons - Nature
    Mar 2, 2021 · Srivastava, D. & DeWitt, N. In vivo cellular reprogramming: the next generation. Cell 166, 1386–1396 (2016). Article CAS PubMed PubMed Central ...
  102. [102]
  103. [103]
    Cardiac regeneration by direct reprogramming in this decade ... - NIH
    Jul 1, 2021 · Direct cardiac reprogramming was a pioneering discovery when it was first reported in 2010, but the cardiomyocyte induction efficiency at the ...
  104. [104]
    In vivo conversion of astrocytes to neurons in the injured adult spinal ...
    Feb 25, 2014 · Here we show that resident astrocytes can be converted to doublecortin (DCX)-positive neuroblasts by a single transcription factor, SOX2, in the injured adult ...
  105. [105]
    In Vivo Reprogramming Using Yamanaka Factors in the CNS
    This review summarizes the current state of in vivo reprogramming research in the central nervous system, focusing on the use of Yamanaka factors.
  106. [106]
    In Vivo Amelioration of Age-Associated Hallmarks by Partial ...
    ... in vivo cellular reprogramming. These experiments demonstrate that the negative consequences of in vivo reprogramming can be avoided by reducing levels of ...
  107. [107]
    Multi-omics characterization of partial chemical reprogramming ...
    Mar 22, 2024 · Partial reprogramming by cyclic short-term expression of Yamanaka factors holds promise for shifting cells to younger states and ...
  108. [108]
    Partial reprogramming induces a steady decline in epigenetic age ...
    It is well established that partial reprogramming happens within an early, reversible phase during the iPSC reprogramming time‐course, which involves the ...
  109. [109]
    Multi-omics characterization of partial chemical reprogramming ...
    Mar 22, 2024 · Partial reprogramming by cyclic short-term expression of Yamanaka factors holds promise for shifting cells to younger states and ...
  110. [110]
    Gene Therapy-Mediated Partial Reprogramming Extends Lifespan ...
    Feb 15, 2024 · Recent studies have demonstrated that partial reprogramming using the Yamanaka factors (or a subset; OCT4, SOX2, and KLF4; OSK) can reverse age- ...<|separator|>
  111. [111]
    Age reprogramming: cell rejuvenation by partial reprogramming - PMC
    Nov 16, 2022 · Partial reprogramming was shown to induce de-differentiation with suppression of cellular identity and activation of pluripotency genes (even ...
  112. [112]
    Chemical reprogramming ameliorates cellular hallmarks of aging ...
    Jun 30, 2025 · Multiple hallmarks of aging can be ameliorated following partial cellular reprogramming by expression of the Yamanaka factors (OSKM) in vitro ...
  113. [113]
    Lessons Learned from Somatic Cell Nuclear Transfer - MDPI
    However, the consensus is that the low efficiency in creating normal viable offspring in animals by SCNT (1–5%) and the high number of abnormalities seen in ...3. Early Scnt Protocol... · 5. Epigenetic Reprogramming... · 5.3. Improving Scnt With...
  114. [114]
    Comparison of Reprogramming Efficiency Between Transduction of ...
    Aug 18, 2010 · Cell–cell fusion-mediated reprogramming may be faster and more efficient than the iPSC method, because the hESC used for cell fusion provides ...
  115. [115]
    [PDF] Single-Cell Expression Analyses during Cellular Reprogramming ...
    Of note is that the variability between single cells in early time points was masked in nonclonal cell populations, as detected by qRT-PCR. (Figure 3D). To ...
  116. [116]
    Epigenetic predisposition to reprogramming fates in somatic cells
    Jan 19, 2015 · Our findings emphasize the relevance of preexisting cell‐to‐cell variability in reprogramming, expanding prior studies that pointed to the early ...
  117. [117]
    Proliferation Rate of Somatic Cells Affects Reprogramming Efficiency
    Our data demonstrated that the proliferation rate of the somatic cell plays critical roles in reprogramming. Slowing down the proliferation of the original ...
  118. [118]
    Systematic assessment of variability in the proteome of iPSC ...
    Protocols generating multiple cell types can lead to higher variability due to differences in cell type ratios. Furthermore, different cell types also express ...
  119. [119]
    Two Supporting Factors Greatly Improve the Efficiency of Human ...
    Nov 6, 2008 · In summary, we have identified two factors, p53 siRNA and UTF1, that can increase the efficiency of human iPSC generation by up to 100 times in ...
  120. [120]
    Differentiation success of reprogrammed cells is heterogeneous in ...
    Apr 8, 2025 · In summary, our study reveals substantial variability in reprogramming efficiency across cell types and identifies the inappropriate expression ...
  121. [121]
    Pluripotent Stem Cell-Based Cell Therapy—Promise and Challenges
    Oct 1, 2020 · Tumorigenicity Caused by Reprogramming Factors. This risk is specific for iPSCs. All of the four reprogramming factors have been associated with ...
  122. [122]
    Prevention of tumor risk associated with the reprogramming of ... - NIH
    In this review, we discuss the current understanding of the risk of tumor formation associated with the reprogramming of various human stem cell-like cells and ...
  123. [123]
    Application of the Yamanaka Transcription Factors Oct4, Sox2, Klf4 ...
    Aug 26, 2023 · Reprogramming generates newly differentiated cells for potential therapies in cancer, neurodegenerative diseases, and rejuvenation processes. In ...
  124. [124]
    Limiting replication stress during somatic cell reprogramming ...
    Aug 21, 2015 · The source of genomic instability on iPSC remains unresolved, although several evidence suggest that it could be linked to replication stress ( ...
  125. [125]
    Tracing genomic instability in induced mesenchymal stromal cell ...
    Apr 14, 2025 · Several studies have reported small genomic alterations in iPS cell lines, including CNAs and SNVs, which may reflect the mutagenic effects of ...
  126. [126]
    Genomic instability in induced stem cells | Cell Death & Differentiation
    Feb 11, 2011 · However, p53 is a DNA damage response gene, raising the possibility that reprogramming is accompanied by DNA damage and genomic instability.
  127. [127]
    The p53–PUMA axis suppresses iPSC generation - Nature
    Jul 22, 2013 · Our results demonstrate that PUMA is a limiting factor for the reprogramming of iPSCs through induction of apoptosis and expectedly genomic ...
  128. [128]
    Elimination of tumorigenic pluripotent stem cells from their ...
    Jul 8, 2025 · This clearly demonstrates that the risk of tumorigenicity is not an imaginary threat and could occur in any patient receiving iPSC-derived ...
  129. [129]
    Tumorigenicity risk of iPSCs in vivo: nip it in the bud - PMC - NIH
    Additionally, the heterogeneity of the cellular product derived from iPSCs for clinical therapy could lead to potential tumorigenicity risks in vivo.
  130. [130]
    Prevention of tumor risk associated with the reprogramming of ...
    Jun 3, 2020 · In this review, we discuss the current understanding of the risk of tumor formation associated with the reprogramming of various human stem ...
  131. [131]
    Cellular plasticity in reprogramming, rejuvenation and tumorigenesis
    Aug 28, 2023 · We discuss how the activity of pioneer TFs generates cellular plasticity that may be critical for inducing not only reprogramming and regeneration, but also ...
  132. [132]
    Epigenetic memory in induced pluripotent stem cells - Nature
    Jul 19, 2010 · Here we demonstrate that iPSCs retain an epigenetic memory of their tissue of origin, which is not apparent when adult somatic cells are ...
  133. [133]
    Epigenetic memory in induced pluripotent stem cells - PubMed - NIH
    Sep 16, 2010 · Such an 'epigenetic memory' of the donor tissue could be reset by differentiation and serial reprogramming, or by treatment of iPSCs with ...
  134. [134]
    Incomplete reprogramming of DNA replication timing in induced ...
    Jan 8, 2024 · The replication timing alterations we identified suggest that some iPSCs may be incompletely reprogrammed or have newly acquired epigenetic ...
  135. [135]
    Epigenetic Memory: Lessons From iPS Cells Derived From Human β ...
    Jan 18, 2021 · Incomplete reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) may be responsible for the heterogeneity in ...
  136. [136]
    H1FOO-DD promotes efficiency and uniformity in reprogramming to ...
    May 2, 2024 · Heterogeneity among both primed and naive pluripotent stem cell lines remains a major unresolved problem. Here we show that express-.
  137. [137]
    Molecular Obstacles to Clinical Translation of iPSCs: Cell Stem Cell
    Jul 21, 2016 · Therefore, incomplete or aberrant reprogramming is mostly a technical restriction rather than a feature intrinsic to the generation of hiPSCs.
  138. [138]
    Induced Pluripotent Stem Cells (iPSCs)—Roles in Regenerative ...
    Since the controversy around ethicality of ESCs continue to be debated, iPSCs have been used to circumvent the process around destruction of the human embryo.
  139. [139]
    Ethics and Induced Pluripotent Stem Cells
    Jun 10, 2009 · The use of iPSCs as an alternative to ESCs may eliminate both the health risks to the donor and the issues of appropriate compensation, as ...
  140. [140]
    Two sides of the same coin? Unraveling subtle differences between ...
    Nov 28, 2017 · The discovery of iPSCs, as patient-specific PSCs, has overcome both of these ethical and immunological issues, making iPSCs attractive ...
  141. [141]
    Safer stem cells for therapy - Harvard Gazette
    Jun 26, 2009 · “The main advantage of iPS cells is that they can be derived from any living individual, especially patients. Pluripotent embryonic stem cells, ...
  142. [142]
    The Promise and Challenge of Induced Pluripotent Stem Cells for ...
    The authors review the basic concept of induction of pluripotency as a novel approach to understand cardiac regeneration, cardiovascular disease modeling, and ...
  143. [143]
    ESCs vs. iPSCs: Which Offers Better Path to Pluripotency?
    Aug 14, 2023 · iPSCs offer the advantage of being patient-specific, bypassing issues of immune rejection. They provide a valuable tool for modeling diseases ...
  144. [144]
    Research and therapy with induced pluripotent stem cells (iPSCs)
    Nov 21, 2019 · Despite the ethical advantage of iPSCs over ESCs, there are still significant concerns regarding the ability of iPSCs to be used for production ...
  145. [145]
    Stem Cell Quick Reference - Learn Genetics Utah
    iPS cells are much less expensive to create than ES cells generated through therapeutic cloning (another type of patient-specific stem cell; see below). However ...<|separator|>
  146. [146]
    Embryonic stem cell and induced pluripotent stem cell - Nature
    Dec 18, 2012 · They can divide indefinitely in vitro, while maintaining the capacity to generate all the cell types of an adult organism.
  147. [147]
    Comparison of Human Induced Pluripotent and Embryonic Stem Cells
    Human induced pluripotent stem cells (hiPSCs) have been hailed as an effective replacement for human embryonic stem cells (hESCs) and a prime candidate cell ...
  148. [148]
    Embryonic stem cells or induced pluripotent stem cells? A DNA ...
    Embryonic stem cells (ESCs), which can theoretically differentiate into any cell type, have become a key cellular tool in biology and have paved the way for ...
  149. [149]
    promises and realities of induced pluripotent stem cells - PubMed
    In 2006, Yamanaka's group announced they had discovered the proverbial "fountain of youth" for human cells, forever changing the field of stem cell research.
  150. [150]
    Primer and interviews: Promises and realities of induced pluripotent ...
    Jul 14, 2011 · This primer discusses those hurdles, iPSC basics, and alternative translational applications that promise to revolutionize medical science.
  151. [151]
    Ten years of progress and promise of induced pluripotent stem cells
    May 11, 2018 · The discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka in 2006 was heralded as a major breakthrough of the decade in stem cell research.
  152. [152]
    California Prop 14 - Stem Cell Bond - SPUR
    Prop 14 authorizes $5.5 billion in bonds for stem cell research, with $1.5 billion for brain/nervous system diseases, and changes CIRM operations.
  153. [153]
  154. [154]
    Human inducible pluripotent stem cells: Realization of initial promise ...
    Sep 2, 2021 · The biopharmaceutical industry has slowly adopted human inducible pluripotent stem cell (hiPSC) technology to enable the development of humanized model systems.Missing: unfulfilled | Show results with:unfulfilled
  155. [155]
    Scaling up pluripotent stem cell-based therapies - ScienceDirect.com
    However, clinical translation of these therapies has been slow, and the field still faces many challenges. There are currently no commercially licensed iPSC- ...
  156. [156]
    Clinical translation of human iPSC technologies: advances, safety ...
    Six major limitations affect the clinical translation of induced pluripotent stem cells (iPSCs): (1) epigenetic reprogramming errors that can introduce ...
  157. [157]
    Pluripotent stem-cell-derived therapies in clinical trial: A 2025 update
    Jan 25, 2025 · The translation of hPSC products into the clinic has been challenged by several technological and regulatory hurdles, which have made hPSC ...
  158. [158]
    Hurdles to clinical translation of human induced pluripotent stem cells
    Stem cell researchers who are trying to bring cell therapies to the clinic face significant translational challenges that by necessity involve a long and costly ...
  159. [159]
    Source cell-type epigenetic memory persists in induced pluripotent ...
    Feb 12, 2024 · We examined the extent of epigenetic and transcriptomic memory persisting first during the transition from differentiated source cell types to iPSCs.
  160. [160]
    Epigenetic Memory and Preferential Lineage-Specific Differentiation ...
    Jul 8, 2011 · Our results suggest that the epigenetic memory may predispose BiPSCs to differentiate more readily into insulin producing cells.
  161. [161]
    [PDF] Epigenetic memory in induced pluripotent stem cells - Harvard DASH
    Such an “epigenetic memory” of the donor tissue could be reset by differentiation and serial reprogramming, or by treatment of iPSC with chromatin-modifying ...
  162. [162]
    A critical view on transgenerational epigenetic inheritance in humans
    Jul 30, 2018 · Transgenerational epigenetic inheritance refers to the transmission of epigenetic information through the germline.
  163. [163]
    Transgenerational epigenetic inheritance: a critical perspective
    Transgenerational epigenetic inheritance in humans and other vertebrates has been controversial for over 150 years and remains so.Introduction · Environmental effects on the... · Darwinian transgenerational...
  164. [164]
    Environmental Impact on Direct Neuronal Reprogramming In Vivo in ...
    Cell reprogramming involves extensive epigenetic modifications, and therefore, the complex in vivo environment is likely to have substantial influences on the ...
  165. [165]
    Transgenerational epigenetic programming via sperm microRNA ...
    Oct 19, 2015 · We demonstrate that sperm miRs function to reduce maternal mRNA stores in early zygotes, ultimately reprogramming gene expression in the offspring hypothalamus.
  166. [166]
    Stochastic biological system-of-systems modelling for iPSC culture
    Jan 8, 2024 · As stem cells are highly sensitive to environmental conditions, a critical concern for iPSC aggregate cultures is spatial heterogeneity.<|control11|><|separator|>
  167. [167]
    Can cell-cultured meat from stem cells pave the way for sustainable ...
    Despite its prominence in global protein production, pig farming is associated with environmental impacts such as greenhouse gas emissions, nitrogen emissions, ...
  168. [168]
    Application of induced pluripotent stem cells in the conservation of ...
    May 28, 2025 · This review synthesizes advances in reprogramming somatic cells from endangered species into iPSCs through integration-free strategies.<|control11|><|separator|>
  169. [169]
    A comparison of non-integrating reprogramming methods - PMC - NIH
    Dec 1, 2014 · The main advantages of the RNA method are the speed of colony emergence, high efficiency, a complete absence of integration, a very low ...
  170. [170]
    Generation of iPSCs by Nonintegrative RNA-Based Reprogramming ...
    Jun 19, 2019 · In this study, 2 different RNA-based footprint-free methods for the generation of iPSCs were compared: the use of synthetic modified messenger ...
  171. [171]
    Induced pluripotent stem cells: Generation methods and a ... - Frontiers
    mRNA technology provides a safer reprogramming procedure than other non-integrative delivery methods, especially because the half-life of RNA molecules is short ...
  172. [172]
    Highly efficient reprogramming to pluripotency and directed ...
    Nov 5, 2010 · The study uses synthetic modified mRNA to reprogram human cells to pluripotency and direct differentiation into myogenic cells, with high ...
  173. [173]
    Review An insight into non-integrative gene delivery approaches to ...
    Feb 20, 2019 · Non-integrative gene delivery approaches do not modify the host genome and therefore can generate transgene-free iPSCs.
  174. [174]
    Non-viral methods for generating integration-free, induced ... - PubMed
    Induced pluripotent stem (iPS) cells were created from mouse fibroblasts by induced expression of Yamanaka factors, Oct3/4, Sox2, Klf4, and c-Myc.
  175. [175]
    Transient non-integrative expression of nuclear reprogramming ...
    Mar 24, 2020 · Here we show that transient expression of nuclear reprogramming factors, mediated by expression of mRNAs, promotes a rapid and broad amelioration of cellular ...
  176. [176]
    Why mRNA Reprogramming Is the Key to Safer and Faster iPSC ...
    Jun 10, 2025 · The emergence of mRNA reprogramming offers a precise, efficient, and xeno-free approach to generating induced pluripotent stem cells (iPSCs).
  177. [177]
    Pluripotent Stem Cells: Recent Advances and Emerging Trends - PMC
    Mar 21, 2025 · Modern reprogramming methods have reduced genomic alterations through the development of safer non-integrative approaches, including messenger ...
  178. [178]
    mRNA-based technology for engineered regenerative medicine
    Aug 26, 2025 · Compared with DNA-based reprogramming methods, mRNA-mediated reprogramming offers a transient, integration-free strategy, reducing genomic ...
  179. [179]
    History and current status of clinical studies using human pluripotent ...
    Apr 6, 2023 · In the common allogeneic settings, there is no advantage obtained when using iPSCs compared with ESCs.
  180. [180]
    Clinical translation of human iPSC technologies: advances, safety ...
    Summary of clinical trials involving induced pluripotent stem cell (iPSC)-based therapies. The table outlines trial name and institution, iPSC-derived cell ...
  181. [181]
    Two-year safety outcomes of iPS cell-derived mesenchymal stromal ...
    May 22, 2024 · We now report results at the 2-year follow-up: 9 of 15 (60%) participants survived, which compares favorably with previously reported outcomes ...Missing: regenerative | Show results with:regenerative
  182. [182]
    A new kind of stem cell is revolutionizing regenerative medicine
    Apr 1, 2025 · Researchers report that their experimental induced pluripotent stem cell, or iPSC, therapeutics could restore vision and cure Type 1 diabetes.
  183. [183]
    The Challenges to Advancing Induced Pluripotent Stem Cell ... - NIH
    Jan 5, 2024 · Challenges include safety, potency, genetic stability, immunogenicity, tumorgenicity, cell reproducibility, scalability, engraftment, and local ...
  184. [184]
    Chemically induced reprogramming to reverse cellular aging - NIH
    Jul 12, 2023 · In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat Aging. 2022; 2:243–53. 10.1038 ...
  185. [185]
    Gene Therapy-Mediated Partial Reprogramming Extends Lifespan ...
    In this study, we show that systemically delivered adeno-associated viruses, encoding an inducible OSK system, in 124-week-old male mice extend the median ...
  186. [186]
    Loss of Epigenetic Information Can Drive Aging, Restoration Can ...
    Jan 12, 2023 · Genetics professor David Sinclair explains how changes to DNA organization and regulation can accelerate or reverse signs of aging in mice.Missing: partial | Show results with:partial
  187. [187]
    Epigenetic reprogramming as a key to reverse ageing and increase ...
    These mechanisms ensure proper development, tissue maintenance, and response to environmental cues throughout an organism's lifespan (Kane and Sinclair, 2019).
  188. [188]
    Life Bio ready for world's first partial epigenetic reprogramming trials
    Mar 13, 2025 · Back in 2020, a seminal paper in Nature showed that treatment with a partial epigenetic reprogramming restored vision in blind mice. Since then, ...
  189. [189]
    Current advances and future prospects of cell reprogramming in ...
    Feb 18, 2025 · Cell reprogramming consists in the reverse process to cell differentiation, making cells lose their identity and age-related characteristics.Abstract · Current state of cell... · New approaches for cell... · Conclusion