Fact-checked by Grok 2 weeks ago

Cell potency

Cell potency denotes the developmental potential of a cell to differentiate into specialized cell types, a fundamental concept in biology and embryogenesis. are characterized by their ability to self-renew and generate differentiated progeny, with potency levels ranging from totipotent, capable of forming an entire including extra-embryonic tissues, to unipotent, restricted to a single . Totipotent cells, exemplified by the zygote and early blastomeres of the morula stage, possess the broadest potency, enabling the formation of all embryonic and extra-embryonic structures such as the placenta. Pluripotent stem cells, including those derived from the inner cell mass of the blastocyst, can differentiate into cells of the three primary germ layers—ectoderm, mesoderm, and endoderm—but lack the ability to produce extra-embryonic tissues. Multipotent stem cells, such as hematopoietic stem cells in bone marrow, are lineage-restricted and can generate multiple but limited cell types within a specific tissue or organ system. Potency is assessed through functional assays including teratoma formation, chimera integration, and differentiation, alongside molecular markers like OCT4, , and NANOG expression. This hierarchy underpins embryonic development, where progressive restriction of potency drives tissue specification during and . In , understanding and manipulating cell potency enables applications such as generation for disease modeling and potential therapies, though challenges persist in achieving stable, high-fidelity differentiation.

Definition and Fundamentals

Core Principles of Differentiation Potential

Cell differentiation potential, commonly referred to as potency, quantifies the range of specialized cell types a progenitor or stem cell can produce through lineage commitment and maturation. This capacity originates at the totipotent stage in the zygote and initial blastomeres, which can generate all embryonic cell lineages as well as extraembryonic tissues such as the placenta and trophoblast. Potency then diminishes progressively during embryogenesis, reflecting a core principle of developmental hierarchy where cells transition from broad versatility to restricted fates, ensuring organized tissue formation without ectopic differentiation. A foundational principle is the unidirectional restriction of potential, driven by causal interactions between intrinsic genetic programs and extrinsic microenvironmental signals. In pluripotent cells, such as those derived from the inner cell mass of the blastocyst, core transcription factors including OCT4, SOX2, and NANOG maintain an undifferentiated state by activating self-renewal pathways and suppressing differentiation-inducing genes across all three germ layers (ectoderm, mesoderm, endoderm). Differentiation initiates via signaling cascades—such as fibroblast growth factor (FGF), bone morphogenetic protein (BMP), and Wnt pathways—that asymmetrically activate lineage-specific transcription factors (e.g., SOX17 for endoderm, TBXT for mesoderm), progressively extinguishing alternative potentials. This process is empirically observed in vitro through directed differentiation assays, where pluripotent cells yield multipotent progenitors limited to tissue-specific subtypes, such as hematopoietic stem cells capable of producing only blood lineage cells (e.g., erythrocytes, leukocytes). Epigenetic remodeling constitutes another key principle, establishing stable, heritable barriers to potency reversal. Undifferentiated cells exhibit bivalent chromatin domains, marked by concurrent active (H3K4me3) and repressive (H3K27me3) histone modifications on developmental loci, enabling rapid activation upon signaling. Upon differentiation, these resolve into lineage-locked states via DNA hypermethylation of pluripotency genes and hypo-methylation of somatic enhancers, rendering reversion improbable under physiological conditions—as evidenced by the failure of most differentiated cells to dedifferentiate spontaneously, even in regenerative contexts like planarian neoblasts. This causal realism underscores potency as an emergent property of molecular determinism, where loss of flexibility supports multicellular complexity, with empirical validation through single-cell epigenomic profiling showing irreversible chromatin compaction in committed progenitors. Multipotent and lower potency levels (oligopotent, unipotent) further exemplify this, confining output to subsets or single types within a lineage, as in unipotent spermatogonial stem cells yielding only sperm.

Hierarchy and Classification of Potency Levels

Cell potency is hierarchically classified based on the extent of differentiation potential, reflecting a progressive restriction from broad developmental versatility to specialized commitment. This classification encompasses five primary levels—totipotent, pluripotent, multipotent, oligopotent, and unipotent—ordered by decreasing capacity to generate diverse cell types, as observed in embryonic and adult contexts. The hierarchy aligns with developmental biology principles, where early embryonic cells exhibit maximal potency, which diminishes through asymmetric division, epigenetic silencing, and lineage-specific gene activation, ultimately yielding terminally differentiated cells. Experimental validation of potency relies on assays such as teratoma formation for pluripotency or chimera integration for totipotency, though these methods underscore variability in potency states rather than absolute categories. Totipotent cells represent the apex, capable of differentiating into all embryonic cell lineages plus extraembryonic tissues like the and , sufficient to form a complete . Such potency is transient, observed in the and cleavage-stage blastomeres up to the 2- to 4-cell stage in mammals, after which cells lose extraembryonic potential. Rare experimental induction of totipotency in cultured cells, such as via overexpression of specific factors in embryonic stem cells, has been reported but remains inefficient and context-dependent. Pluripotent cells follow, able to produce all somatic cell types derived from the three germ layers—ectoderm, mesoderm, and endoderm—but not extraembryonic structures. This level is exemplified by embryonic stem cells derived from the inner cell mass of blastocysts around 4-5 days post-fertilization in humans, and induced pluripotent stem cells reprogrammed from somatic cells using factors like Oct4, Sox2, Klf4, and c-Myc, as established in 2006. Pluripotency is maintained in vitro through culture on feeder layers or with leukemia inhibitory factor, but spontaneous differentiation highlights its metastable nature. Multipotent cells possess restricted potential to differentiate into multiple, but lineage-limited, cell types within a particular tissue or organ system, such as hematopoietic stem cells yielding erythrocytes, leukocytes, and platelets from bone marrow. These are prevalent in adult tissues, including neural stem cells in the subventricular zone capable of generating neurons, astrocytes, and oligodendrocytes, with potency assayed via colony-forming units or transplantation reconstitution. Unlike pluripotent cells, multipotent ones exhibit quiescence and self-renewal tailored to tissue homeostasis rather than organismal development. Oligopotent cells further narrow the scope, differentiating into a small subset of cell types within a multipotent lineage, such as common myeloid progenitors that yield macrophages, granulocytes, and erythrocytes but not lymphoid cells. This level bridges multipotency and unipotency, often representing committed progenitors in processes like hematopoiesis, where potency is quantified by single-cell clonal assays showing limited output diversity. Unipotent cells at the base exhibit the narrowest potency, self-renewing and differentiating solely into one mature cell type, as seen in spermatogonial stem cells producing spermatozoa or basal epidermal cells generating . Despite limited , unipotent stem cells sustain lifelong renewal, with their potency confirmed by tracing and lack of multi-lineage potential in transplantation studies. This hierarchy is not rigidly discrete; transitional states and , such as , challenge strict boundaries, particularly under experimental manipulation.
Potency LevelKey Differentiation CapacityCanonical Examples
TotipotentEntire organism (embryonic + extraembryonic)Zygote, 2-cell blastomeres
PluripotentThree germ layers (somatic only)Embryonic stem cells, iPSCs
MultipotentMultiple types in one lineageHematopoietic stem cells, mesenchymal stem cells
OligopotentFew types in a sub-lineageMyeloid progenitors
UnipotentSingle mature cell typeSpermatogonia, epidermal progenitors

Historical Development

Pre-20th Century Observations

Early observations of regeneration in simple organisms suggested that certain tissues possessed cells capable of extensive and self-renewal. In 1744, Abraham Trembley demonstrated that freshwater polyps () could regenerate complete organisms from small fragments, including the formation of multiple heads from a single bisected specimen, challenging preformationist views and implying inherent developmental versatility in their cellular components. This phenomenon indicated a form of totipotency at the tissue level, as severed parts reorganized into fully functional individuals without external templates. Building on such findings, Lazzaro Spallanzani's experiments in 1768 on amphibians revealed limb regeneration in salamanders and newts, where amputated tails or legs reformed through a proliferative mass of tissue—later termed the —that differentiated into bone, muscle, and skin. Spallanzani noted the blastema's role in recapitulating embryonic-like processes, with varying regenerative capacity across species; for instance, frogs regenerated tails but lost this ability post-metamorphosis, while mammals showed limited repair via scarring rather than patterned regrowth. These studies highlighted species-specific differences in cellular potency, foreshadowing hierarchical potentials in vertebrate development. In embryology, Caspar Friedrich Wolff's 1759 dissertation advanced epigenesis, proposing that organs arise progressively from an initially homogeneous, fluid-like substance through solidification and differentiation, rather than preformed miniatures unfolding. Wolff's microscopic observations of chick embryos supported this, showing sequential formation of structures like the intestine from presumptive tissues, establishing a framework for understanding cellular specialization from undifferentiated precursors. This contrasted with dominant preformation theories and emphasized causal processes in potency realization. The consolidation of cell theory in the mid-19th century further illuminated potency constraints. in 1838 described plant cells as arising from nucleated sacs, while in 1839 extended this to animals, positing cells as the universal building blocks formed by crystallization-like processes. Rudolf Virchow's 1858 axiom "omnis cellula e cellula" underscored that cells derive from preexisting cells, implying lineage-restricted differentiation potentials rather than . These insights shifted focus to cellular and variability in developmental capacity. Ernst Haeckel introduced the term Stammzelle (stem cell) in 1868, initially in an evolutionary context to denote primordial unicellular ancestors capable of giving rise to multicellular lineages through division and specialization. Though phylogenetic in origin, the concept soon influenced embryological interpretations of cells retaining broad differentiative potential, bridging regeneration and development observations. Such pre-20th century work laid empirical groundwork for later potency classifications, revealing cells' variable abilities to generate diverse progeny amid debates over mechanism and inheritance.

20th Century Milestones in Stem Cell Identification

Early theoretical foundations for stem cell identification emerged in the hematology field during the first decade of the 20th century. In 1909, Alexander A. Maximow proposed the unitarian theory of hematopoiesis, positing a common multipotent precursor cell in bone marrow as the origin of all blood cell lineages, based on comparative histological studies of mesenchyme-derived tissues across species.00019-7) This concept, building on earlier observations by figures like Artur Pappenheim, laid groundwork for recognizing stem-like cells but lacked experimental validation of self-renewal or clonal differentiation. Definitive experimental identification of hematopoietic stem cells (HSCs) occurred in 1961 through the work of James E. Till and Ernest A. McCulloch at the Cancer Institute. By transplanting cells from donor mice into lethally irradiated recipients and observing discrete nodules (colonies) in the , they demonstrated that individual cells could proliferate and differentiate into multiple blood lineages, evidenced by the clonal nature confirmed in subsequent cytological analyses. Their colony-forming unit (CFU-S) quantified these multipotent, self-renewing cells, with sensitivity studies showing a small subpopulation resistant to doses that killed differentiated progenitors, thus establishing HSCs as quiescent, long-term repopulating cells capable of multilineage reconstitution. This milestone provided the first functional proof of with restricted potency, enabling transplantation therapies. A pivotal advance in identifying pluripotent stem cells came in 1981 with the derivation of embryonic stem cell (ESC) lines from mouse preimplantation embryos. Martin J. Evans and Matthew H. Kaufman isolated pluripotential cells from the inner cell mass of blastocysts, maintaining them in undifferentiated culture using feeder layers and serum, and demonstrated their capacity to form teratocarcinomas containing derivatives of all three germ layers upon injection into adult mice. Concurrently and independently, Gail R. Martin established similar lines from 3.5-day mouse embryos cultured in medium conditioned by splenic factors, coining the term "embryonic stem cells" to distinguish them from teratocarcinoma-derived lines; her assays included morphological criteria and differentiation into embryoid bodies exhibiting neural, muscular, and epithelial tissues. These cells exhibited high nucleocytoplasmic ratios, alkaline phosphatase activity, and the ability to contribute to all tissues in chimeras, confirming pluripotency defined by broad differentiation potential without totipotency. The 20th century closed with the isolation of human embryonic stem cells in 1998 by James A. Thomson and colleagues at the University of Wisconsin. Using blastocysts from in vitro fertilization surplus, they derived stable lines maintained on mouse feeders with basic fibroblast growth factor, verifying pluripotency through spontaneous differentiation into trophoblasts, neurons, and cardiomyocytes, as well as teratoma formation in immunodeficient mice showing derivatives from ectoderm, mesoderm, and endoderm. This achievement extended mouse ESC protocols to primates (earlier derivations in rhesus monkeys by the same group in 1995), enabling human-specific potency studies while highlighting ethical considerations in sourcing. These milestones shifted stem cell research from theoretical and multipotent adult types to experimentally tractable pluripotent sources, underpinning later potency classifications.

21st Century Breakthroughs in Reprogramming

In 2006, Shinya Yamanaka and colleagues demonstrated that mouse fibroblasts could be reprogrammed to an induced pluripotent state by ectopic expression of four transcription factors: Oct4, Sox2, Klf4, and c-Myc, enabling the cells to form teratomas and contribute to chimeric mice with germline transmission.00976-7) This breakthrough established that somatic cells retain epigenetic information sufficient for reversal to pluripotency without nuclear transfer, challenging prior assumptions of irreversible differentiation. The method's efficiency was initially low, around 0.01-0.1%, and relied on retroviral vectors, raising concerns over insertional mutagenesis. By 2007, Yamanaka's team extended the approach to human fibroblasts, generating human iPSCs that expressed pluripotency markers and differentiated into all three germ layers, while James Thomson independently identified a similar factor set (Oct4, Sox2, Nanog, Lin28) for human cells. These human iPSCs mirrored embryonic stem cells in morphology, gene expression, and epigenetic profiles, but initial protocols carried risks of tumorigenesis due to c-Myc's oncogenicity and viral integration. Yamanaka received the 2012 Nobel Prize in Physiology or Medicine, shared with John Gurdon, for these discoveries validating reprogramming's feasibility across species. Subsequent refinements addressed safety and scalability. Non-integrating methods, such as virus vectors (2008) and episomal plasmids (2009), minimized genomic alterations, achieving integration-free iPSCs with efficiencies up to 0.001% but improved purity. Small-molecule cocktails, including inhibitors of deacetylases and TGF-β signaling, boosted reprogramming efficiency to over 10-fold by 2010, facilitating partial epigenetic erasure without full factor reliance. By 2014, Yamanaka's group produced the first clinical-grade human iPSCs using defined feeder-free conditions and xeno-free media, enabling autologous therapies while reducing immunogenicity risks. Direct lineage reprogramming emerged as a complementary advance, bypassing pluripotency for targeted transdifferentiation; for instance, fibroblasts were converted to neurons (2010) or cardiomyocytes (2010) using lineage-specific factors, offering faster, safer paths for tissue repair without tumorigenic intermediates. Chemical-only reprogramming protocols, devoid of genetic material, generated iPSC-like cells from mouse fibroblasts by 2013, though human applications lagged due to incomplete pluripotency validation. These developments expanded potency manipulation's scope, with over 1,000 iPSC lines banked globally by 2018 for disease modeling and drug screening. Despite progress, challenges persist in maturation fidelity and scalability, with clinical trials for macular degeneration (e.g., Japan's 2014 autologous retinal transplant) highlighting variable efficacy.

Totipotency

Defining Features and Mechanisms

Totipotent cells are defined by their ability to differentiate into every cell type of an organism, including embryonic lineages and extraembryonic tissues such as trophoblast and primitive endoderm that contribute to placental structures. This capacity extends beyond pluripotency, which is limited to somatic cell types, by enabling the formation of a complete, organized body plan culminating in a fertile adult. The zygote represents the archetypal totipotent cell, as fertilization equips it with the coordinated developmental program necessary for autonomous organismal development upon implantation. In mammals, totipotency manifests transiently during early embryogenesis, persisting through the one-cell (zygote) and two-cell stages in mice, and up to the four-cell stage in humans, after which blastomeres lose independent potential to generate extraembryonic derivatives. Experimental validation of totipotency relies on functional assays, such as isolating blastomeres and assessing their ability to develop into blastocysts with both inner cell mass and trophectoderm compartments, or transferring them to pseudopregnant hosts to yield viable offspring. These criteria distinguish totipotent cells from pluripotent ones, which fail to organize extraembryonic contributions or sustain full embryonic development. Molecular mechanisms underpinning totipotency involve profound epigenetic reconfiguration following fertilization, characterized by genome-wide DNA demethylation, active and passive, mediated by factors like Stella (Dppa3) that protect against deleterious retrotransposon derepression. Zygotic genome activation (ZGA), initiated by pioneer transcription factors such as Dux in mice or DUX4 in humans, drives expression of totipotency-associated genes including Zscan4 and establishes non-canonical histone marks like H3K4me3 at promoter regions devoid of CpG islands. This reconfiguration, coupled with maternal mRNA clearance and histone variant deposition (e.g., H3.3), creates a permissive chromatin landscape for broad lineage priming. Totipotency is lost progressively as cells divide, through dilution of transient maternal factors, accumulation of lineage-restrictive epigenetic marks, and activation of signaling pathways that bias toward pluripotency in the . In vitro, totipotent-like states can be induced in embryonic stem cells via chemical or genetic perturbations, yielding two-cell-like cells (2CLCs) with transcriptomic profiles mirroring early embryos and enhanced chimerism potential, as demonstrated by efficiency improvements with treatment.

Natural Occurrences and Experimental Induction

In mammalian development, totipotency manifests naturally in the zygote, the single cell formed immediately following fertilization, which possesses the complete genetic and epigenetic potential to generate an entire organism, encompassing both embryonic lineages and extra-embryonic tissues such as the placenta and trophoblast. This capacity arises from the fusion of haploid gametes, enabling symmetric cleavage divisions that initially preserve developmental equivalence among daughter cells. In mice, totipotency extends to individual blastomeres of the two-cell and four-cell embryos, as demonstrated by their ability to form viable chimeric offspring or complete blastocysts when isolated and cultured, though potency wanes by the eight-cell stage due to asymmetric divisions and lineage restrictions. Human totipotency similarly persists through the zygote and early cleavage stages up to the eight-cell embryo, after which cells transition toward restricted potentials, reflecting conserved mechanisms across eutherian mammals despite species-specific timings. Experimental induction of totipotency has proven challenging, as it requires not only reprogramming cellular identity but also recapitulating the unique zygotic transcriptome, epigenome, and bidirectional differentiation capacity absent in later stem cell types like embryonic stem cells (ESCs), which are pluripotent. Somatic cell nuclear transfer (SCNT), pioneered in frogs in 1952 and achieved in mammals with Dolly the sheep in 1996, represents an early method to induce totipotency by injecting a differentiated somatic nucleus into an enucleated oocyte, triggering remodeling to a zygote-like state capable of full embryonic development. More recent chemical approaches have advanced this frontier; for instance, in 2022, a defined cocktail of three small molecules (including inhibition of histone deacetylases and activation of specific signaling pathways) converted mouse ESCs into totipotent-like cells expressing zygotic genes like Zscan4 and capable of forming both embryonic and extra-embryonic structures in chimeras. Further innovations include spliceosomal modulation, where pharmacological inhibition of splicing factors in human pluripotent stem cells (hPSCs) in 2024 induced a totipotent state marked by blastomere-like morphology, oscillatory gene expression, and formation of trophectoderm-extraembryonic endoderm structures, though sustained maintenance remains limited.00519-1) Overexpression of specific factors, such as Hmgn3 in mouse ESCs, has also triggered totipotency-associated markers and enhanced chimeric contribution, including placental tissues, as reported in 2025 studies linking it to chromatin remodeling. These methods, while promising for modeling preimplantation development, face hurdles in stability and human applicability, with induced totipotent cells often exhibiting transient rather than heritable potency, underscoring the causal primacy of early embryonic cues like retinoic acid signaling in natural totipotency windows. Ongoing research prioritizes orthogonal validation through tetraploid complementation assays, which rigorously test for extra-embryonic potential beyond teratoma formation used for pluripotency.

Pluripotency

Embryonic Sources and Characteristics

Embryonic pluripotent stem cells are derived from the (ICM) of blastocyst-stage embryos, which form approximately 4-5 days after fertilization in . These blastocysts typically consist of 100-200 cells, with the ICM comprising about 20-30 pluripotent cells surrounded by the trophectoderm. Human embryonic cells (hESCs) are isolated by immunosurgery or of the ICM, followed by on feeder layers or in defined media to promote outgrowth and establishment of self-renewing cell lines. The first successful derivation of hESC lines occurred in from surplus embryos generated via fertilization. hESCs exhibit indefinite self-renewal capacity in vitro, proliferating while maintaining an undifferentiated state under specific culture conditions involving basic fibroblast growth factor (FGF2) and inhibitors of differentiation pathways. Pluripotency is evidenced by their ability to differentiate into derivatives of all three germ layers—ectoderm, mesoderm, and endoderm—both in vitro through embryoid body formation or directed protocols, and in vivo via teratoma formation in immunocompromised mice, which contain tissues representative of multiple lineages. Key molecular hallmarks include expression of transcription factors such as OCT4, SOX2, and NANOG, which form a regulatory network sustaining the pluripotent ground state, alongside surface markers like SSEA-4, TRA-1-60, and TRA-1-81. Human ESCs typically reside in a "primed" epigenetic state, distinct from the "naïve" state of mouse ESCs, characterized by higher X-chromosome inactivation in female lines and reliance on activin/Nodal signaling for maintenance. These cells demonstrate high activity, enabling replicative immortality without immediate , and maintain a stable diploid in early passages, though long-term culture can lead to genetic instability or adaptations favoring over differentiation fidelity. Unlike totipotent cells, hESCs cannot contribute to extra-embryonic tissues like or primitive , limiting their developmental potential to post-implantation equivalents.

Induced Pluripotency and Reprogramming

Induced pluripotency refers to the artificial reprogramming of differentiated somatic cells into a pluripotent state resembling that of embryonic stem cells (ESCs), enabling self-renewal and differentiation into all three germ layers. This process was first achieved in 2006 by Kazutoshi Takahashi and Shinya Yamanaka, who introduced four specific transcription factors—Oct4 (also known as Oct3/4), Sox2, Klf4, and c-Myc—into mouse embryonic and adult fibroblasts via retroviral vectors, generating cells termed induced pluripotent stem cells (iPSCs) that formed teratomas and contributed to chimeric mice.00976-7) The reprogramming efficiency was low, initially around 0.01-0.1%, reflecting the stochastic nature of the process involving epigenetic barriers and somatic memory. In 2007, the same Yamanaka factors successfully reprogrammed human adult dermal fibroblasts into iPSCs, which exhibited ESC-like morphology, gene expression, and differentiation potential, including teratoma formation in immunodeficient mice.01471-7) Independently, James Thomson's group generated human iPSCs using a slightly different combination: Oct4, Sox2, Nanog, and Lin28, avoiding oncogenic c-Myc to reduce tumorigenicity risks. These transcription factors act as pioneers, binding to closed chromatin, initiating a cascade that silences lineage-specific genes while activating pluripotency networks, such as the Oct4-Sox2-Nanog circuit. The molecular mechanisms of reprogramming involve coordinated changes: transcriptional activation of pluripotency genes, epigenetic remodeling (e.g., DNA demethylation and histone modifications to erase somatic imprints), metabolic shifts toward glycolysis, and suppression of somatic signaling pathways like TGF-β. Yamanaka factors collectively drive these events, with Oct4 and Sox2 coregulating pluripotency enhancers, Klf4 counteracting differentiation cues, and c-Myc promoting proliferation but contributing to genomic instability. Despite advances like non-integrating delivery methods (e.g., Sendai virus or mRNA) to minimize mutagenesis, challenges persist, including incomplete reprogramming leading to biased differentiation, persistent epigenetic abnormalities, and tumorigenic potential from residual pluripotency or insertional mutations. Subsequent optimizations have improved safety and efficiency, such as omitting c-Myc to lower sarcoma risk, though at the cost of reduced reprogramming rates, and exploring chemical cocktails or protein transduction to avoid genetic integration entirely.00214-8) These iPSCs demonstrate that adult cell potency can be experimentally reset, bypassing ethical concerns of embryonic sources while enabling patient-specific modeling, though clinical translation requires rigorous validation for genetic fidelity and functional equivalence to ESCs.

Functional States and Transitions

Pluripotent stem cells exhibit distinct functional states, primarily the naive and primed states, which correspond to developmental stages in the early embryo. The naive state mirrors the pre-implantation inner cell mass (ICM) of the blastocyst, featuring homogeneous self-renewal, dome-shaped colony morphology in mouse embryonic stem cells (ESCs), and dependency on leukemia inhibitory factor (LIF)/STAT3 signaling for maintenance. In contrast, the primed state resembles the post-implantation epiblast, with cells showing flattened colonies, reliance on fibroblast growth factor 2 (FGF2) and Activin/Nodal signaling, and a poised epigenetic landscape for lineage commitment. These states differ in transcriptional profiles, epigenetic modifications, and developmental potential; naive cells display lower X-chromosome inactivation (XCI) dosage compensation, bivalent chromatin domains with reduced H3K27me3 at developmental genes, and broader enhancer activation compared to primed cells, which exhibit primed XCI and more restricted enhancer usage.00140-4) Naive pluripotency supports efficient chimera formation and germline transmission in mice, whereas primed cells are biased toward neuroectoderm differentiation and show limited chimerism. In humans, conventional embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are predominantly primed, though protocols have enabled derivation of naive-like human pluripotent stem cells (hPSCs) exhibiting expanded potency, including trophectoderm potential.00158-2) Transitions between states occur naturally during mammalian , progressing from naive to primed pluripotency upon implantation, driven by downregulation of LIF/ and upregulation of FGF/ERK signaling, alongside epigenetic remodeling such as XCI and enhancer reorganization. 00140-4) In vitro, primed-to-naive conversion in human PSCs involves chemical cocktails inhibiting GSK3β, MEK/ERK, PKC, and pathways, often combined with LIF, Activin, and inhibition, yielding cells with transcriptomes approximating pre-implantation epiblast.00158-2) Reverse transitions from naive to primed can be induced by FGF2 supplementation, mimicking implantation cues, though naive states are metastable and prone to reversion without precise signaling . These conversions highlight the of pluripotency, governed by dynamic signaling networks and transitions, with implications for modeling peri-implantation .

Validation and Assay Methods

Validation of pluripotency in stem cells requires functional assays that demonstrate the capacity to generate differentiated derivatives representing all three embryonic germ layers—ectoderm, mesoderm, and endoderm—alongside molecular and epigenetic characterizations. These methods distinguish truly pluripotent cells from those with partial or restricted potential, as marker expression alone, such as OCT4, NANOG, and SOX2, is insufficient without functional proof. The teratoma formation assay serves as the established gold standard for assessing pluripotency in human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). In this in vivo method, 1–10 million undifferentiated cells are injected subcutaneously or intratesticularly into immunocompromised mice, such as NOD/SCID strains, where they proliferate and differentiate into teratomas containing ectodermal (e.g., neural tissue), mesodermal (e.g., cartilage, muscle), and endodermal (e.g., gut epithelium) structures, confirmed via histological analysis after 8–12 weeks. The assay's sensitivity allows detection of residual undifferentiated cells in differentiated populations, though it is time-consuming, variable due to host factors, and raises tumorigenicity concerns, prompting refinements like standardized protocols to improve reproducibility. Chimera formation assays provide a stringent functional test, particularly in models, by injecting pluripotent cells into preimplantation blastocysts and evaluating their contribution to all fetal tissues, including , in resulting chimeric . For cells, ethical constraints limit homologous chimeras, leading to interspecies approaches, such as injecting hPSCs into morulae or epiblasts, where integration and multi-lineage contribution indicate pluripotency, though efficiency remains low and species barriers hinder robust validation. In vitro assays complement in vivo methods by enabling rapid, scalable assessment without animal use. Directed differentiation protocols induce hPSCs to form lineage-specific progenitors or mature cells from each germ layer—e.g., SOX17+ FOXA2+ endoderm, TBXT+ mesoderm, and PAX6+ ectoderm—verified by immunofluorescence, qPCR, or flow cytometry for markers like brachyury or nestin. Embryoid body formation, where aggregates spontaneously differentiate into multi-lineage structures mimicking early embryogenesis, offers a simpler alternative, though less directed. The International Stem Cell Initiative's comparative analysis of over 150 hPSC lines confirmed that while in vitro assays predict differentiation potential reliably, they correlate less strongly with full pluripotency than teratoma formation, underscoring the need for integrated approaches. Additional validation includes genomic integrity checks (e.g., karyotyping, SNP arrays) to rule out abnormalities, and epigenetic profiling (e.g., DNA methylation at promoter regions of pluripotency genes) to confirm naive or primed states, but these support rather than replace functional assays. Ongoing efforts aim to refine assays for higher throughput and ethical compliance, such as advanced in vitro organoid models, yet teratoma and chimera tests remain benchmarks for causal demonstration of pluripotency.

Restricted Potency Types

Multipotency in Adult Tissues

Multipotent stem cells in adult tissues possess the ability to self-renew and differentiate into a limited range of cell types within specific lineages, contrasting with the broader potential of pluripotent cells. These cells maintain tissue homeostasis and support repair in mature organisms, primarily through lineage-restricted differentiation pathways verified by in vitro colony-forming assays and in vivo transplantation studies. For instance, long-term hematopoietic stem cells (LT-HSCs) in bone marrow sustain lifelong blood production by generating all myeloid and lymphoid lineages. Hematopoietic stem cells exemplify multipotency, as evidenced by their capacity to repopulate irradiated recipients with multilineage progeny, including erythrocytes, megakaryocytes, granulocytes, and lymphocytes. Functional assays, such as competitive repopulation in mouse models, demonstrate that LT-HSCs maintain this potency over extended periods, with clonal tracking revealing strong erythroid bias in some multipotent subsets under stress conditions. In humans, retroviral marking studies confirm the multipotent nature of these cells, supporting their hierarchical role in steady-state and regenerative hematopoiesis. Mesenchymal stem cells (MSCs), derived from bone marrow stroma, adipose tissue, and other adult sources, differentiate into osteoblasts, chondrocytes, and adipocytes, fulfilling criteria for multipotency within mesodermal lineages. Isolation protocols yield MSCs at frequencies of approximately 0.001-0.01% in bone marrow, with trilineage potential confirmed via directed differentiation and gene expression analyses. Clinical evidence from autologous transplants further validates their role in bone and cartilage repair, though potency varies by donor age and tissue source. Neural stem cells in the adult mammalian brain, located in the and hippocampal subgranular zone, generate neurons, , and , as shown by lineage tracing and neurosphere assays. These cells persist throughout adulthood, contributing to at rates of about 700 new neurons daily in the human , with multipotency restricted to glial and neuronal fates. Transplantation and clonal analyses underscore their self-renewal and regional specificity, though their regenerative output diminishes with .

Oligopotency and Unipotency Examples

Oligopotent cells possess a restricted differentiation potential, capable of giving rise to only a small number of related cell types while retaining self-renewal capacity. In hematopoiesis, common lymphoid progenitors (CLPs) exemplify oligopotency by differentiating into B lymphocytes, T lymphocytes, and natural killer (NK) cells, thereby supporting adaptive and innate immune functions within the lymphoid lineage. Similarly, common myeloid progenitors (CMPs) demonstrate oligopotency through their ability to produce granulocytes, macrophages, megakaryocytes, and erythrocytes, facilitating key aspects of innate immunity, platelet formation, and oxygen transport. These progenitors arise from multipotent hematopoietic stem cells in the bone marrow and commit progressively to narrower lineages under the influence of specific transcription factors and cytokines. Unipotent stem cells exhibit the most limited potency, self-renewing to produce progeny that differentiate exclusively into one mature cell type. Spermatogonial stem cells in the testes represent a classic unipotent example, undergoing continuous division to generate spermatozoa while maintaining the germline pool throughout adult life. Epidermal basal stem cells in the skin interfollicular epidermis also embody unipotency, proliferating to yield keratinocytes that form the protective stratum corneum barrier against environmental stressors. In skeletal muscle, satellite cells function as unipotent progenitors, activating post-injury to fuse and repair myofibers by producing myoblasts that differentiate solely into multinucleated muscle fibers. Although some debate exists regarding the existence of truly unipotent stem cells due to potential latent multipotency under experimental conditions, these examples align with observed lineage restriction in vivo, where differentiation is tightly regulated by niche signals and epigenetic barriers.

Assessment and Technological Advances

Traditional In Vivo and In Vitro Assays

Traditional in vivo assays for assessing cell potency primarily evaluate the functional differentiation potential of injected cells within a living organism. For pluripotent stem cells, the teratoma formation assay, established as a gold standard since the derivation of human embryonic stem cells in 1998, involves subcutaneous or intratesticular injection of 1–10 million undifferentiated cells into immunodeficient mice, such as SCID or NOD-SCID strains; tumors typically form within 6–12 weeks and are histologically examined for the presence of differentiated tissues representative of all three germ layers—ectoderm (e.g., neural tissue), mesoderm (e.g., cartilage, muscle), and endoderm (e.g., gut epithelium)—to confirm pluripotency. The chimera formation assay, a cornerstone for mouse embryonic stem cells since the 1980s, entails injecting 10–15 pluripotent cells into a blastocyst-stage embryo, which is then implanted into a pseudopregnant female; potency is gauged by the donor cells' contribution to chimeric tissues across lineages, with germline transmission (e.g., coat color or genetic markers in offspring) providing the most stringent evidence of full pluripotency.30213-2) For multipotent cells, such as hematopoietic stem cells (HSCs), the competitive repopulation assay—developed in the 1970s—involves transplanting candidate cells mixed with wild-type bone marrow into lethally irradiated recipient mice; multilineage engraftment (e.g., >1% contribution to myeloid, lymphoid, and erythroid compartments over 4–6 months, often via serial transplantation) demonstrates self-renewal and multipotency. Similarly, the CFU-S (colony-forming unit-spleen) assay, pioneered by Till and McCulloch in 1961, injects cells intravenously into irradiated mice, where splenic nodules form after 8–12 days, revealing proliferative potential and limited differentiation into erythroid, myeloid, or megakaryocytic lineages. In vitro assays offer controlled, ethical alternatives or complements to in vivo methods, focusing on self-organized or induced differentiation in culture. For pluripotency validation, the embryoid body (EB) assay, routine since the 1990s for embryonic stem cells, suspends dissociated cells in low-attachment plates or hanging drops to form three-dimensional aggregates (typically 100–500 μm diameter after 2–5 days), followed by adhesion and spontaneous differentiation over 10–21 days; potency is assessed by expression of lineage-specific markers (e.g., Nestin for ectoderm, α-smooth muscle actin for mesoderm, α-fetoprotein for endoderm) via immunofluorescence, RT-PCR, or flow cytometry, though it lacks the organismal integration of in vivo tests. For restricted potency, colony-forming unit (CFU) assays, foundational since the 1960s for hematopoietic progenitors, seed single cells or low-density populations in semi-solid methylcellulose media supplemented with cytokines (e.g., erythropoietin, stem cell factor); colonies (50–500 cells after 7–14 days) are morphologically classified—such as CFU-GM for granulocyte-macrophage multipotency or burst-forming unit-erythroid (BFU-E) for erythroid commitment—to quantify proliferative and differentiation capacity. In mesenchymal stem/stromal cells, the fibroblast colony-forming unit-fibroblast (CFU-F) assay plates cells at clonal density (e.g., 10–100 cells/cm²) in basal medium; colonies (>50 cells after 10–14 days) are stained (e.g., with crystal violet) and evaluated for multilineage potential via subsequent adipogenic, osteogenic, or chondrogenic induction. These assays, while quantitative and scalable, often underestimate long-term self-renewal compared to in vivo counterparts due to the absence of physiological niches.00114-8)

Modern Tools for Potency Evaluation

Single-cell RNA sequencing (scRNA-seq) has emerged as a pivotal tool for evaluating stem cell potency by quantifying transcriptional heterogeneity and estimating differentiation potential at the individual cell level. Techniques such as signaling entropy analysis, applied to datasets from over 1,000 cells, correlate transcriptional noise with pluripotency, where higher entropy indicates greater potency in embryonic stem cells compared to differentiated states. Similarly, intrinsic dimensionality metrics derived from scRNA-seq data enable direct potency scoring without reliance on lineage tracing, revealing potency gradients in human pluripotent stem cells (hPSCs) through manifold learning algorithms. These methods outperform traditional marker-based assays by capturing population heterogeneity, as demonstrated in analyses of naive and primed hPSC states where scRNA-seq identified modular transcriptional programs linked to potency transitions. CRISPR-Cas9-based functional genomics screens provide high-throughput validation of potency by perturbing candidate genes and assessing impacts on self-renewal and differentiation in hPSCs. Genome-scale CRISPR knockout screens in human iPSCs have identified core chromatin regulators, such as those in the Polycomb repressive complex, essential for maintaining pluripotent identity while decoupling it from proliferation fitness. Activation screens using CRISPRa further quantify enhancer efficacy and gene regulatory networks influencing potency, with barcoded reporters showing that basal expression and open chromatin predict editing outcomes in hPSCs. These assays, conducted in 2023-2024, enable causal inference on potency determinants, surpassing correlative transcriptomics by directly testing gene function in relevant cellular contexts. Epigenomic profiling assays, including ATAC-seq for chromatin accessibility and bisulfite sequencing for DNA methylation, complement transcriptomic tools by mapping the regulatory landscape underlying potency states. In hPSCs, these reveal dynamic remodeling during reprogramming, where naive pluripotency exhibits hypomethylated promoters and accessible enhancers distinct from primed states, correlating with differentiation bias. Integrated multi-omics approaches, combining scRNA-seq with epigenomics, have quantified potency via epigenetic entropy, identifying biomarkers for stable pluripotency in iPSC lines derived post-2020. Such tools facilitate rapid, non-destructive evaluation, essential for quality control in therapeutic-grade stem cell production. Machine learning-enhanced analyses of omics data streamline potency assessment by predicting functional outcomes from high-dimensional profiles. For instance, correlation-based metrics like CCAT integrate connectome and transcriptome data to score single-cell potency scalably, validated on datasets exceeding 10,000 cells with accuracy rivaling in vivo chimeras. These computational frameworks, refined in 2020-2025 studies, reduce reliance on animal models while accounting for biases in source-derived iPSCs, though they require validation against gold-standard assays for controversial potency claims.

Applications and Impacts

Regenerative Medicine and Therapy Development

Human pluripotent stem cells (hPSCs), encompassing embryonic stem cells and induced pluripotent stem cells, exploit their capacity to differentiate into derivatives of all three germ layers to produce transplantable cell populations for treating degenerative diseases. These cells enable the generation of functional tissues such as retinal pigment epithelium for age-related macular degeneration, dopaminergic neurons for Parkinson's disease, and cardiomyocytes for myocardial infarction, addressing limitations of donor organ shortages and immune incompatibility in traditional transplants. Induced pluripotent stem cells (iPSCs), reprogrammed from somatic cells, facilitate autologous therapies that minimize rejection risks, as demonstrated in Japan's 2014 clinical trial transplanting iPSC-derived retinal cells into patients with wet macular degeneration, where cells integrated without tumor formation after one year of follow-up. Clinical translation has accelerated, with 115 regulatory-approved trials for 83 distinct hPSC-derived products registered worldwide as of December 2024, primarily targeting ophthalmic, neurological, and cardiovascular conditions. Early-phase trials report favorable safety profiles, including no ectopic differentiation or uncontrolled proliferation in most cases, though efficacy varies; for instance, hESC-derived retinal implants improved visual acuity in some dry macular degeneration patients in a 2020 U.S. trial, with sustained effects observed up to two years post-transplantation. For cardiac repair, iPSC-derived cardiomyocytes injected post-myocardial infarction in preclinical models reduced scar size by 40-50% and enhanced ejection fraction by 10-15 percentage points, prompting phase I/II human trials initiated in 2023 that confirm engraftment without arrhythmias in initial cohorts. Multipotent adult stem cells, such as hematopoietic stem cells, underpin established therapies like bone marrow transplants for leukemia, where high-potency progenitors reconstitute the blood system in over 80% of matched-donor cases annually worldwide. Emerging strategies combine potency modulation—e.g., partial reprogramming to enhance multipotency—with bioengineering scaffolds to regenerate complex organs; a 2024 study using iPSC-derived progenitors seeded on decellularized matrices yielded functional mini-kidneys that restored partial renal function in rodent models of chronic kidney disease. Scaling manufacturing remains critical, with advances in bioreactor systems achieving 10^9-scale production of GMP-compliant iPSC derivatives by 2025, supporting broader trial enrollment.00678-4/fulltext) Therapy development also integrates potency assays to ensure differentiated cells retain therapeutic fidelity, reducing risks like teratoma formation from residual undifferentiated cells, which occurred in <1% of monitored trial participants. Ongoing efforts focus on allogeneic "off-the-shelf" banks of universal iPSCs edited via CRISPR to ablate HLA antigens, potentially slashing costs by 50-70% compared to autologous approaches while maintaining potency. These innovations position cell potency as foundational to regenerative paradigms, with projected market expansion to $4.7 billion for iPSC therapies by 2033, driven by successes in vision and diabetes reversal trials.

Disease Modeling and Research Utilities

Induced pluripotent stem cells (iPSCs), derived by reprogramming somatic cells to a pluripotent state, enable patient-specific disease modeling by differentiating into diverse cell types relevant to particular pathologies. This approach recapitulates disease phenotypes , such as neuronal aggregates exhibiting inclusions in models from patient-derived iPSCs differentiated into neurons. For monogenic disorders like Timothy syndrome, iPSCs harboring the CACNA1C produce cardiomyocytes displaying irregular beating and calcium handling defects, mirroring clinical arrhythmias.00209-9) Pluripotent stem cells facilitate high-fidelity models of complex, polygenic diseases inaccessible in animal systems, including amyotrophic lateral sclerosis (ALS), where motor neurons from iPSC lines reveal TDP-43 aggregation and hyperexcitability as early pathogenic events.00007-8) In cardiovascular research, iPSC-derived cardiomyocytes from long QT syndrome patients exhibit prolonged action potentials, enabling mechanistic studies of ion channel dysfunction. These models support causal inference by isolating genetic variants' effects in human cellular contexts, bypassing interspecies translational gaps observed in rodent models. Beyond modeling, cells of varying potencies underpin drug discovery pipelines through phenotypic screening and toxicity assays. iPSC-derived hepatocytes and cardiomyocytes predict idiosyncratic drug-induced liver injury and cardiotoxicity with higher human relevance than immortalized cell lines, as demonstrated in screens identifying hERG channel blockers. Multipotent mesenchymal stem cells (MSCs), restricted to mesodermal lineages, model tissue-specific inflammatory responses, such as in osteoarthritis, where their osteogenic differentiation reveals paracrine signaling defects in diseased states. Hematopoietic stem cells, oligopotent progenitors, inform leukemia modeling by propagating patient mutations in erythro-myeloid lineages for targeted therapy validation. These utilities extend to high-throughput platforms, where CRISPR-edited iPSCs test variant pathogenicity, accelerating precision medicine; for instance, editing SCN5A mutations in iPSC-cardiomyocytes has validated loss-of-function mechanisms in Brugada syndrome. However, variability in differentiation efficiency and epigenetic memory from reprogramming sources can confound reproducibility, necessitating standardized protocols. Overall, potency hierarchies—pluripotent for broad recapitulation, multipotent for lineage fidelity—complement each other in dissecting disease causality and therapeutic responses.

Challenges, Risks, and Debates

Technical Limitations and Safety Concerns

Technical limitations in manipulating cell potency primarily stem from inefficiencies in reprogramming somatic cells to induced pluripotent states, where success rates remain low, often below 1% even with optimized protocols involving factors like Oct4, Sox2, Klf4, and c-Myc. Incomplete reprogramming leads to epigenetic memory, wherein induced pluripotent stem cells (iPSCs) retain chromatin marks and DNA methylation patterns from their somatic origins, biasing subsequent differentiation toward the donor tissue type rather than enabling unbiased pluripotency. This memory persists despite apparent pluripotent markers and can only be partially erased through serial reprogramming or differentiation-reprogramming cycles, limiting the versatility of iPSCs compared to embryonic stem cells (ESCs). Genomic instability represents another core limitation, with iPSCs exhibiting higher rates of chromosomal aberrations, copy number variations, and point mutations than ESCs, often arising during the reprogramming process due to oxidative stress, DNA damage response deficiencies, and reactivation of transposable elements. Studies indicate that up to 20-30% of iPSC lines harbor detectable genetic alterations, including recurrent gains at chromosome 20q11.21 associated with proliferative advantages, which undermine long-term stability and potency fidelity. Differentiation protocols for assessing or harnessing potency suffer from variability, with inconsistent yields and heterogeneous cell populations, complicating scalable production for therapeutic use. Safety concerns are dominated by the tumorigenic potential of pluripotent cells, as undifferentiated ESCs or iPSCs can form teratomas—benign tumors containing derivatives of all three germ layers—upon transplantation, with risks persisting if even trace residual pluripotent cells evade purification. In preclinical models, teratoma incidence correlates with pluripotency markers like Oct4 expression, necessitating stringent sorting techniques such as fluorescence-activated cell sorting, yet no method guarantees complete elimination without compromising yield. Genomic instability exacerbates this, as acquired mutations may confer malignant transformation, with reports of iPSC-derived tumors showing oncogenic signatures akin to sarcomas or carcinomas. Immune compatibility issues further heighten risks, as autologous iPSCs can elicit aberrant responses due to reprogramming-induced neoantigens or incomplete MHC matching. These factors collectively demand rigorous preclinical validation, including karyotyping and whole-genome sequencing, to mitigate clinical translation hazards. The derivation of human embryonic stem cells (hESCs), first achieved in 1998 by James Thomson's team at the University of Wisconsin-Madison, necessitates the destruction of early-stage embryos, typically blastocysts at the 5- to 8-cell stage, sparking profound ethical debates over the moral status of the embryo. Opponents, including bioethicists and pro-life advocates, contend that such embryos represent nascent human life deserving protection, equating their destruction to a form of homicide, while proponents emphasize the potential to alleviate suffering from degenerative diseases like Parkinson's and spinal cord injuries. This tension culminated in policy restrictions, such as the U.S. federal funding ban on new hESC lines announced by President George W. Bush on August 9, 2001, which limited support to 21 existing lines deemed non-controversial, a policy reversed by President Barack Obama on March 9, 2009, amid arguments that ethical oversight could mitigate concerns without halting research. The advent of induced pluripotent stem cells (iPSCs) in 2006, pioneered by through adult s using four transcription factors (Oct4, , , and c-Myc), offered an ethically preferable alternative by bypassing use, earning Yamanaka the 2012 Nobel Prize in Physiology or Medicine shared with . Despite this, iPSCs face scrutiny over incomplete epigenetic , potential genomic instability, and incomplete equivalence to hESCs in potency, with studies indicating retained "epigenetic memory" biasing toward the donor cell type and elevated risks of tumorigenesis like formation upon transplantation. Ethically, while iPSCs avoid embryo destruction, concerns persist regarding for somatic cell donors, especially in autologous therapies, and the possibility of use in some variants, though these pale compared to hESC sourcing.00276-9) Source-related controversies extend to credibility biases in reporting and institutional priorities, where academic and media outlets, often aligned with progressive funding agendas, have disproportionately hyped hESC potential while underreporting successes in adult multipotent stem cells, which have yielded over 1,000 clinical trials by 2020 with tangible outcomes in conditions like hematopoietic reconstitution, versus fewer hESC-derived therapies reaching approval. Peer-reviewed analyses highlight how mainstream media coverage from 2000-2010 favored embryonic approaches by a 4:1 ratio, framing ethical opposition as ideologically driven rather than philosophically grounded, potentially skewing public and policy perceptions despite empirical evidence of adult stem cells' superior safety profile in avoiding immune rejection without teratoma risks. Such biases, rooted in institutional incentives for high-impact pluripotent research, underscore the need for skepticism toward consensus narratives in stem cell potency claims, as evidenced by retracted high-profile studies like the 2014 STAP cell scandal claiming stimulus-induced pluripotency from differentiated cells.

Regulatory Hurdles and Future Prospects

Regulatory approval for therapies derived from high-potency cells, such as pluripotent stem cells (PSCs), faces stringent oversight from agencies like the U.S. Food and Drug Administration (FDA), which classifies them as somatic cellular therapies requiring demonstration of safety, purity, potency, and efficacy through phased clinical trials. Key hurdles include ensuring genetic stability and minimizing tumorigenicity risks, as undifferentiated PSCs can form teratomas upon transplantation, necessitating rigorous purification protocols and long-term preclinical data. Scalability and manufacturing reproducibility pose additional challenges, with variations in cell differentiation efficiency complicating batch-to-batch consistency and increasing costs for good manufacturing practice (GMP)-compliant production. Immunogenicity and engraftment issues further complicate approval, particularly for allogeneic -derived products, where immune rejection risks demand advanced or autologous approaches like induced pluripotent s (iPSCs), though the latter's reprogramming introduces potential off-target mutations. As of December 2024, only 115 interventional human PSC (hPSC) trials had received regulatory approval worldwide, predominantly in early phases (Phase I/II), reflecting delays due to these technical and safety barriers rather than outright bans. Ethical regulations add layers, with FDA prohibitions on editing in therapies to prevent heritable changes, alongside requirements for addressing therapeutic misconception in trial participants. Looking ahead, integration of gene editing tools like CRISPR-Cas9 with iPSCs promises enhanced potency control and reduced , enabling precise lineage-specific for organoid-based therapies targeting diseases like Parkinson's and . Advances in off-the-shelf allogeneic iPSCs, combined with immune evasion strategies, could accelerate scalability and lower costs, with early 2025 reports indicating successful vision restoration and Type 1 diabetes reversal in preclinical models. Regulatory , including FDA's streamlined pathways for regenerative medicines, may shorten approval timelines, fostering broader clinical adoption by 2030, provided long-term efficacy data from ongoing trials confirm durable engraftment without adverse events. Precision medicine approaches tailoring cell potency to patient hold potential to transform regenerative applications, though sustained investment in standardization remains essential to overcome persistent reproducibility hurdles.

References

  1. [1]
    Describing the Stem Cell Potency: The Various Methods of ...
    Stem cells are defined by their capabilities to self-renew and give rise to various types of differentiated cells depending on their potency.
  2. [2]
    Basics of Stem Cell Biology as Applied to the Brain - NCBI - NIH
    Jul 27, 2016 · Not all stem cells have the same “potency,” the capacity to give rise to similar cell types.Introduction · The Basic Biology of Stem Cells · How to Test Pluripotency?<|separator|>
  3. [3]
    Cell Potency: Totipotent vs Pluripotent vs Multipotent Stem Cells
    Jul 31, 2025 · Cell potency refers to the varying ability of stem cells to differentiate into specialized cell types. Cells with the greatest potency can ...
  4. [4]
    Stem cells: a comprehensive review of origins and emerging clinical ...
    Aug 25, 2022 · The ability of stem cells to differentiate into multiple cell types is termed potency, and stem cells can be classified by their potential ...
  5. [5]
    From pluripotency to totipotency: an experimentalist's guide to ...
    Summary: This Primer refines the term totipotency, explores the nature of cells reported to be totipotent and discusses the experimental capacity of these.Missing: hierarchy | Show results with:hierarchy
  6. [6]
    [PDF] Teacher Background Stem Cells - OHSU
    Stem cells can be classified by their potential to differentiate. Totipotent and pluripotent describe embryonic stem cells. Multipotent, oligopotent, unipotent ...Missing: hierarchy | Show results with:hierarchy
  7. [7]
    Hydra, a fruitful model system for 270 years - PubMed
    The discovery of Hydra regeneration by Abraham Trembley in 1744 promoted much scientific curiosity thanks to his clever design of experimental strategies ...
  8. [8]
    Abraham Trembley's Approach to Research on the Organism—1744 ...
    While studying hydra, Trembley demonstrated that: (a) complete animals can regenerate from small, cut pieces of those animals; (b) animals can reproduce ...
  9. [9]
    Lazzaro Spallanzani | Italian Physiologist & Microbiologist | Britannica
    The results of his regeneration and transplantation experiments appeared in 1768. ... His transplantation experiments showed great experimental skill and included ...
  10. [10]
    Caspar Friedrich Wolff's Theory of Epigenesis - jstor
    In his dissertation, Wolff proposed a model for development in plants and animals based on two factors: the ability of plant and animal fluids to solidify, and ...
  11. [11]
    1 - Pre-Kantian Revival of Epigenesis: Caspar Friedrich Wolff's De ...
    Mar 10, 2023 · According to the German embryologist Caspar Friedrich Wolff (1733–94), the body cannot be conceived without the existence of a epigenetic force.
  12. [12]
    "The Cell-Theory" (1853), by Thomas Henry Huxley
    Dec 12, 2013 · The twenty-two page article reviews twelve works on cell theory, including those in Germany by Caspar Friedrich Wolff in the eighteenth century ...
  13. [13]
    From haematopoietic stem cells to complex differentiation landscapes
    Jun 7, 2019 · When Ernst Haeckel first used the word stem cell (Stammzelle) in 1868, as a Darwinist he used it to refer to the primordial unicellular ...
  14. [14]
    Ambiguous Cells: The Emergence of the Stem Cell Concept in the ...
    The stem cell concept emerged in late 19th century Germany, with Boveri's work around 1900, and was applied in embryology and blood work, with both positive ...
  15. [15]
    100 plus years of stem cell research—20 years of ISSCR
    Jun 14, 2022 · This review looks back at some of the key developments in stem cell research as well as the evolution of the ISSCR as part of that field.Missing: potency | Show results with:potency
  16. [16]
    Stem cells: past, present, and future
    Feb 26, 2019 · This paper is a review focused on the discovery of different stem cells and the potential therapies based on these cells.
  17. [17]
    The origins of the identification and isolation of hematopoietic stem ...
    Till and McCulloch proposed that these latter cells were hematopoietic stem cells, that these cells could self-renew, and that they could make all blood cell ...
  18. [18]
    The road ended up at stem cells - PubMed
    The search for hematopoietic stem cells (HSC) actually started with the discovery by Till, McCulloch, and colleagues (1-3) that bone marrow contained single ...
  19. [19]
    Stem cells and their dual properties: self-renewal and differentiation
    Feb 27, 2021 · Till and McCulloch's discoveries explained the basis of bone marrow transplantation, which prolongs the lives of patients with leukemia and ...
  20. [20]
    Establishment in culture of pluripotential cells from mouse embryos
    Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981 Jul 9;292(5819):154-6. doi: 10.1038/292154a0. Authors. M J Evans, M H Kaufman.
  21. [21]
    Isolation of a pluripotent cell line from early mouse embryos cultured ...
    This report describes the establishment directly from normal preimplantation mouse embryos of a cell line that forms teratocarcinomas when injected into mice.
  22. [22]
    Pluripotent stem cells: private obsession and public expectation - PMC
    In 1981 Martin Evans, Matt Kaufman and Gail Martin found that cells from early mouse embryos exposed to the same culture environment can suspend developmental ...
  23. [23]
    Embryonic stem cell lines derived from human blastocysts - PubMed
    Embryonic stem cell lines derived from human blastocysts. Science. 1998 Nov 6;282(5391):1145-7. doi: 10.1126/science.282.5391.1145. Authors. J A Thomson , J ...
  24. [24]
    Embryonic Stem Cell Lines Derived from Human Blastocysts - Science
    James A. Thomson et al. ,. Embryonic Stem Cell Lines Derived from Human Blastocysts.Science282,1145-1147(1998).DOI:10.1126/science.282.5391.1145. Export ...Missing: James | Show results with:James
  25. [25]
    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.
  26. [26]
    Induced pluripotent stem cells (iPSCs): molecular mechanisms of ...
    Apr 26, 2024 · Given the critical role of DNA methylation in establishing epigenetic memory during cell differentiation, active remodeling of DNA methylation ...
  27. [27]
    Induced pluripotent stem cell technology: a decade of progress - PMC
    Since the advent of induced pluripotent stem cell (iPSC) technology a decade ago, enormous progress has been made in stem cell biology and regenerative ...
  28. [28]
    Shinya Yamanaka produces first clinical-grade iPS cells
    Jun 13, 2014 · In 2006, he discovered a way to produce iPS cells without using embryonic stem cells, first with mouse skin cells, and the following year with ...
  29. [29]
    The evolving biology of cell reprogramming - PMC
    Exciting opportunities arise from the process known as 'cellular reprogramming' in which cells can be reliably changed from one tissue type to another.
  30. [30]
    Chemical reprogramming for cell fate manipulation
    Sep 7, 2023 · Chemical reprogramming offers an unprecedented opportunity to control somatic cell fate and generate desired cell types including pluripotent stem cells.
  31. [31]
    Clinical translation of human iPSC technologies: advances, safety ...
    A major breakthrough came in 2006 when Takahashi and Yamanaka identified four transcription factors, OCT4, SOX2, KLF4, and c-MYC (OSKM), capable of ...
  32. [32]
    Totipotency: What It Is and What It Is Not - PMC - NIH
    Consequently, a cell that is totipotent is also a one-cell embryo; that is, a cell that is capable of generating a globally coordinated developmental sequence.
  33. [33]
    Molecular mechanisms underlying totipotency - PMC - NIH
    Sep 4, 2023 · In stem cell research, totipotent-like cells often refer to cells with broader plasticity than pluripotent stem cells. As mouse embryonic stem ...
  34. [34]
  35. [35]
    Retinoic acid signaling is critical during the totipotency window in ...
    May 27, 2021 · Totipotency is a transient feature of the cells in the early embryo, which in mice is limited to the zygote and 2-cell embryo, because only the ...<|control11|><|separator|>
  36. [36]
    Totipotency of mouse zygotes extends to single blastomeres of ...
    May 27, 2021 · We found that totipotent cells are present up to the four-cell stage of mouse embryos, and that pre- and peri-implantation developmental ...Missing: occurrences | Show results with:occurrences
  37. [37]
    Splicing inhibition unlocks human totipotency | Nature Methods
    Aug 9, 2024 · In humans, totipotency is a transient state observed in the zygote through the eight-cell blastomeres, which makes generating and maintaining ...
  38. [38]
    Cell totipotency: molecular features, induction, and maintenance
    The totipotent stage of developing embryos is associated with unique transcriptional and epigenetic states. Understanding the mechanism for generation, ...
  39. [39]
    sufficient to induce totipotency in embryonic stem cells - Nature
    Sep 19, 2022 · Hu et al. 1 report that a cocktail of three small chemicals is sufficient to induce totipotency in murine embryonic stem (ES) cells.
  40. [40]
  41. [41]
    Pluripotent Stem Cells: Current Understanding and Future Directions
    Pluripotent stem cells have the ability to undergo self-renewal and to give rise to all cells of the tissues of the body.
  42. [42]
    Tracing the Derivation of Embryonic Stem Cells from the Inner Cell ...
    May 6, 2010 · The derivation of embryonic stem cells (ESCs) from the inner cell mass (ICM) of mouse blastocysts consisting of about 20 cells occurs in vitro ...
  43. [43]
    Derivation of Embryonic Stem-Cell Lines from Human Blastocysts
    We report the derivation and characterization of 17 additional human embryonic stem cell lines. We obtained frozen cleavage- and blastocyst-stage human embryos.
  44. [44]
    [PDF] Embryonic Stem Cell Lines Derived from Human Blastocysts
    Embryonic Stem Cell Lines. Derived from Human. Blastocysts. James A. Thomson,* Joseph Itskovitz-Eldor, Sander S. Shapiro,. Michelle A. Waknitz, Jennifer J ...
  45. [45]
    Regulation of Embryonic Stem Cell Self-Renewal - PMC
    Human naïve ESCs possess a high degree of plasticity and differentiation potential, thereby providing a novel paradigm for studying early human embryo ...
  46. [46]
    Derivation and Potential Applications of Human Embryonic Stem Cells
    Embryonic stem cells are pluripotent cell lines that are derived from the blastocyst-stage early mammalian embryo. These unique cells are characterized by ...<|separator|>
  47. [47]
    The pluripotency state of human embryonic stem cells derived from ...
    Human embryonic stem cells (hESCs) derived from blastocyst stage embryos present a primed state of pluripotency, whereas mouse ESCs (mESCs) display naïve ...
  48. [48]
    Alterations of proliferative and differentiation potentials of human ...
    It was found that hESCs have a tendency to a higher proliferative ability and a reduced differentiation potential on increasing the passage number. These ...
  49. [49]
    Induction of pluripotent stem cells from mouse embryonic and adult ...
    We demonstrate induction of pluripotent stem cells from mouse embryonic or adult fibroblasts by introducing four factors, Oct3/4, Sox2, c-Myc, and Klf4, under ...
  50. [50]
    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.
  51. [51]
    Perspective Induced Pluripotent Stem Cells: Past, Present, and Future
    Jun 14, 2012 · In 2006, we showed that stem cells with properties similar to ESCs ... Yamanaka, 2006). We designated these cells iPSCs. In 2007, we ...
  52. [52]
    Transcriptional and epigenetic mechanisms of cellular ... - NIH
    Reprogramming of somatic cells into iPSCs requires silencing of the somatic cell transcriptional profile and the induction of an ESC-like expression program ...
  53. [53]
    Cell reprogramming: methods, mechanisms and applications
    Mar 27, 2025 · This review summarizes the recent advances in cell reprogramming mediated by transcription factors or chemical molecules, followed by ...
  54. [54]
    Molecular mechanisms of pluripotency and reprogramming
    Oct 25, 2010 · The four reprogramming factors discovered by Takahashi and Yamanaka, Oct4, Sox2, Klf4 and cMyc, all have vital roles in early embryogenesis and ...Defining Pluripotent Stem... · Inducing Pluripotent Stem... · Molecular Mechanisms Of...<|separator|>
  55. [55]
    The roles of the reprogramming factors Oct4, Sox2 and Klf4 in ...
    Oct 22, 2012 · The most well known of these enhancer factors is c-Myc, which was added alongside O, S and K in the original reprogramming experiment but ...
  56. [56]
    The Challenges to Advancing Induced Pluripotent Stem Cell ... - NIH
    Jan 5, 2024 · iPSC therapies pose significant unique challenges with respect to safety, potency, genetic stability, immunogenicity, tumorgenicity, cell reproducibility, ...Induced Pluripotent Stem... · Autologous Vs Allogeneic... · Engraftment Challenges Of...
  57. [57]
    Pluripotent Stem Cell-Based Cell Therapy—Promise and Challenges
    Oct 1, 2020 · Another critical issue is whether iPSC reprogramming is itself mutagenic. Gore et al. performed whole exome sequencing of human iPSC lines ...Introduction · Tumorigenicity · Immunogenicity
  58. [58]
    Decoding human chemical reprogramming: mechanisms and ...
    Chemical reprogramming, by precisely regulating signaling pathways and epigenetic status, induces human pluripotent stem cells (hCiPSCs) in a controlled ...Review · Highlights · Abstract
  59. [59]
    Overcoming barriers to the clinical utilization of iPSCs
    The second limitation is that many iPSC lines are not authentically pluripotent, as many cell lines inefficiently differentiate into differentiated cell types ...
  60. [60]
    Naive and Primed Pluripotent States - ScienceDirect.com
    Jun 5, 2009 · We propose that two phases of pluripotency can be defined: naive and primed. This distinction extends to pluripotent stem cells derived from embryos or by ...
  61. [61]
    Network Features and Dynamical Landscape of Naive and Primed ...
    The naive state of pluripotency characterizes pluripotent stem cells that are able to differentiate into lineages of all germ layers and the germline and to ...
  62. [62]
    Comparative analysis of naive, primed and ground state ... - Nature
    Apr 12, 2018 · While naive and ground state mESCs have more similar but not identical profiles, primed state mESCs show a very distinct profile. We further ...
  63. [63]
    [PDF] Bridging naïve and primed pluripotency - Stanford University
    The transition from naïve to primed pluripotency is a pivotal step in mammalian development that sets the stage for the generation of germ layers and organs,.
  64. [64]
    Cell fate roadmap of human primed-to-naive transition reveals ...
    Jun 7, 2022 · Our study describes a high-resolution cell roadmap toward naive pluripotency, and provides valuable sources for human blastocyst modeling and early ...
  65. [65]
    Epigenetic Regulation of Transition Among Different Pluripotent ...
    Jul 24, 2019 · Here, we summarized current understanding on epigenetic signatures of naive, primed pluripotent stem cells as well as other totipotent-like ...
  66. [66]
    Assessment of established techniques to determine developmental ...
    May 15, 2018 · The International Stem Cell Initiative compared several commonly used approaches to assess human pluripotent stem cells (PSC).
  67. [67]
    Describing the Stem Cell Potency: The Various Methods ... - Frontiers
    Stem cells are defined by their capabilities to self-renew and give rise to various types of differentiated cells depending on their potency.<|separator|>
  68. [68]
    The teratoma assay: an in vivo assessment of pluripotency - PubMed
    In the teratoma assay, putative pluripotent stem cells (PSCs) are implanted into an immune-compromised mouse where they may proliferate and differentiate to ...
  69. [69]
    Teratoma Assay for Testing Pluripotency and Malignancy of Stem ...
    Feb 15, 2023 · The teratoma assay is still the preferred method to assess the presence of undifferentiated cells in a differentiated cell product for transplantation.<|separator|>
  70. [70]
    Standardization of the Teratoma Assay for Analysis of Pluripotency ...
    The assay could detect residual teratoma forming cells within differentiated hESC populations however its sensitivity was decreased in the presence of ...<|separator|>
  71. [71]
    Contributions of Mammalian Chimeras to Pluripotent Stem Cell ...
    Jul 13, 2016 · Chimeras are widely acknowledged as the gold standard for assessing stem cell pluripotency, based on their capacity to test donor cell lineage potential.
  72. [72]
    Chimeric contribution of human extended pluripotent stem cells to ...
    Apr 15, 2021 · Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in ...
  73. [73]
    Human-Mouse Chimerism Validates Human Stem Cell Pluripotency
    Jan 7, 2016 · Chimera formation, generated by stem cell transplantation to the embryo, is a stringent assessment of stem cell pluripotency.
  74. [74]
    Pluripotency Assays for Stem Cells | Cell Quality
    Pluripotency assays include teratoma, embryoid body, directed differentiation to endoderm, mesoderm, and ectoderm, and the STEMdiff™ Trilineage Differentiation ...
  75. [75]
    Evaluating Strategies to Assess the Differentiation Potential of ...
    Sep 28, 2024 · Pluripotent stem cells have the ability to differentiate into all cells and tissues within the human body, and as a result they are ...
  76. [76]
    Pluripotent stem cell assays: Modalities and applications for ...
    This manuscript provides a review focused on embryonic stem cell-based models and their place within the landscape of alternative developmental toxicity assays.
  77. [77]
    Is normal hematopoiesis maintained solely by long-term multipotent ...
    For more than 50 years now, it has been known that the BM comprises a population of multipotent long-term hematopoietic stem cells (LT-HSCs) that are defined by ...
  78. [78]
    Clonal and Quantitative In Vivo Assessment of Hematopoietic Stem ...
    Mar 21, 2019 · Clonal and Quantitative In Vivo Assessment of Hematopoietic Stem Cell Differentiation Reveals Strong Erythroid Potential of Multipotent Cells.
  79. [79]
    Hematopoietic Stem Cells but Not Multipotent Progenitors Drive ...
    Jun 5, 2018 · We found that HSCs respond differentially to chronic erythroid stress compared with their closely related multipotent progenitors (MPPs).<|separator|>
  80. [80]
    Identification of a Hierarchy of Multipotent Hematopoietic ... - NIH
    MPP have yet to be identified within the human hematopoiesis hierarchy, but functional evidence suggests that they exist. Retroviral or lentiviral marking of ...
  81. [81]
    The role of multipotent marrow stromal cells (MSCs) in tissue ...
    These cells are able to self-renew; they readily expand in culture and can differentiate into mesenchymal cell types, such as cartilage, bone and fat.
  82. [82]
    Human mesenchymal stem cells - current trends and future ...
    Human MSCs (hMSCs) are the non-haematopoietic, multipotent stem cells with the capacity to differentiate into mesodermal lineage such as osteocytes, adipocytes ...<|separator|>
  83. [83]
    Mesenchymal Stem Cells | Circulation Research
    Sep 30, 2011 · Mesenchymal stem cells (MSCs) are a prototypical adult stem cell with capacity for self-renewal and differentiation with a broad tissue distribution.<|separator|>
  84. [84]
    Multipotent CNS Stem Cells Are Present in the Adult Mammalian ...
    The spinal cord and the entire ventricular neuroaxis of the adult mammalian CNS contain multipotent stem cells, present at variable frequency and with unique ...
  85. [85]
    Multipotent neural stem cells originating from neuroepithelium exist ...
    Apr 10, 2025 · In the adult mammalian brain, NSCs exist mainly in two regions: the subgranular zone in the dentate gyrus of the hippocampus and the ...
  86. [86]
    Multipotent CNS Stem Cells Are Present in the Adult Mammalian ...
    Dec 1, 1996 · The spinal cord and the entire ventricular neuroaxis of the adult mammalian CNS contain multipotent stem cells, present at variable frequency and with unique ...
  87. [87]
    Hematopoietic Stem Cells: The Paradigmatic Tissue-Specific Stem ...
    Multipotent progenitors give rise to oligo-potent progenitors including the CLP, which gives rise to mature B lymphocytes, T lymphocytes, and natural killer (NK) ...
  88. [88]
    New paradigms on hematopoietic stem cell differentiation
    Jun 14, 2019 · In this review, we discuss the evidence supporting heterogeneity within HSC and progenitor populations as well as the hierarchical models ...
  89. [89]
    Unipotent - (Cell Biology) - Vocab, Definition, Explanations | Fiveable
    An example of unipotent stem cells is epidermal stem cells, which can only differentiate into keratinocytes, the primary cells of the skin. While unipotent ...
  90. [90]
    Do You Know the 5 Types of Stem Cells? - BioInformant
    Rating 4.8 (86) May 22, 2025 · Specifically, HSCs are oligopotent stem cells that can differentiate into both myeloid and lymphoid cells.
  91. [91]
    Reconsidering pluripotency tests: Do we still need teratoma assays?
    The teratoma assay is used as an in vivo method to test pluripotency of cells, namely the ability of those cells to generate cells/tissue of all three germ ...Missing: traditional | Show results with:traditional
  92. [92]
    Haematopoietic stem cell self-renewal in vivo and ex vivo - PMC - NIH
    This paper describes the use of single cell transplantation assays to identify self-renewing multipotent HSCs within the mouse bone marrow. 31. Waterstrat A ...Hsc Transplantation Assay · Hsc Self-Renewal In Vivo · Allogeneic Hsct
  93. [93]
    CFU-S assay: a historical single-cell assay that offers modern insight ...
    In this review, we explore the major discoveries made with the CFU-S assay, consider its modern use and recent improvements, and compare it with commonly used ...
  94. [94]
    Analysis of Embryoid Bodies Derived from Human Induced ...
    Our paper proposes the embryoid body (EB) assay as a useful in vitro, cost-effective alternate to demonstrate the differentiation potential of derived hiPSCs.
  95. [95]
    Culture and Analysis of Hematopoietic Progenitor Cells in CFU Assays
    The colony-forming unit (CFU) assay is one of the most widely used assays for hematopoietic stem and progenitor cells (HSPCs). CFU assays allow measurement ...
  96. [96]
    Colony forming unit assays for MSCs - PubMed
    This chapter describes 2 methods to assay the colony forming ability of MSCs; (a) a traditional assay for colony forming units (CFUs) and (b) single-cell colony ...
  97. [97]
    Single-cell entropy for accurate estimation of differentiation potency ...
    Jun 1, 2017 · We show that signalling entropy provides a more accurate and robust potency estimate than other entropy-based measures.
  98. [98]
    Intrinsic dimensionality of single-cell transcriptomic data reveals ...
    Jul 24, 2025 · Recent computational advances have enabled estimation of potency directly from single-cell RNA sequencing (scRNA-seq) data, with intrinsic ...
  99. [99]
    Single Cell RNA-Sequencing of Pluripotent States Unlocks Modular ...
    Oct 1, 2015 · Here, we report single cell RNA-sequencing of mESCs cultured in three different conditions: serum, 2i, and the alternative ground state a2i.
  100. [100]
    Parallel genome-scale CRISPR-Cas9 screens uncouple human ...
    Oct 17, 2024 · We further identify a core set of genes controlling both stem cell fitness and pluripotent identity, including a network of chromatin factors.
  101. [101]
    Massively parallel characterization of CRISPR activator efficacy in ...
    Mar 13, 2023 · Analysis of CRISPRa activity in hPSCs with thousands of barcoded reporters. The efficacy of CRISPRa depends on basal expression and chromatin state.
  102. [102]
    CRISPR-based functional genomics screening in human-pluripotent ...
    Apr 18, 2023 · CRISPR-Cas9-based functional genomics screening offers a powerful approach to determine gene function in a high-throughput manner.
  103. [103]
    Epigenetic Control of Cell Potency and Fate Determination during ...
    May 25, 2023 · This review provides an overview of the current understanding of key epigenetic features that underly pluripotency, how this epigenetic landscape is remodelled ...Epigenetic Control Of Cell... · 1. Introduction · 1.3. Epigenetic Remodelling...Missing: assays | Show results with:assays
  104. [104]
    Ultra-fast scalable estimation of single-cell differentiation potency ...
    Nov 27, 2020 · We present a single-cell potency measure, called Correlation of Connectome and Transcriptome (CCAT), which can return accurate single-cell potency estimates of ...
  105. [105]
    Evaluating Strategies to Assess the Differentiation Potential of ...
    Sep 28, 2024 · In this article, we consider the methods used to establish cellular pluripotency and subsequently analyse characterisation data for over 1590 human pluripotent ...
  106. [106]
    Pluripotent Stem Cells: Recent Advances and Emerging Trends - PMC
    Mar 21, 2025 · The paper by Du et al. (2023) examines stem cell-signaling in treating intervertebral disk degeneration, and demonstrates iPSC-derived therapies ...
  107. [107]
    Pluripotent stem-cell-derived therapies in clinical trial: A 2025 update
    Jan 2, 2025 · Here, we review the landscape of interventional hPSC trials worldwide, highlighting available data on clinical safety and efficacy.
  108. [108]
    recent developments and future prospects in stem-cell therapy - PMC
    Nov 5, 2024 · Recent biotechnological advancements, such as exosome-based therapeutics, single-cell RNA sequencing, and CRISPR technology, have revolutionized ...
  109. [109]
    Stem cell-based therapy for human diseases - Nature
    Aug 6, 2022 · This review provides an update on recent clinical applications using either hPSCs or MSCs derived from bone marrow (BM), adipose tissue (AT), or the umbilical ...
  110. [110]
    Combining the induced pluripotent stem cell (iPSC) technology with ...
    Nearly 100 clinical trials using allogeneic CAR-T cells have been designed in recent years, and the released data of some of these trials are promising (Moradi ...
  111. [111]
    iPSC-based cell replacement therapy: from basic research to clinical ...
    This review examines the progression of iPSC-based regenerative medicine, focusing on cell replacement therapy and mechanisms like the Replacement Effect.
  112. [112]
    The Pipeline for of iPSC-Derived Cell Therapeutics in 2025
    Rating 4.9 (45) Sep 30, 2025 · iPSC-derived cells have the potential to be used as therapies for treating cardiovascular, neurological, and metabolic diseases, as well as repairing damaged ...
  113. [113]
    Induced Pluripotent Stem Cells Market Size, Share & Forecast 2025 ...
    Induced Pluripotent Stem Cells Market is expected to reach US$ 4.69 Billion by 2033 from US$ 2.01 Billion in 2024, with a CAGR of 9.86% from 2025 to 2033. The ...
  114. [114]
    Induced pluripotent stem cells in disease modelling and drug ...
    The derivation of induced pluripotent stem cells (iPSCs) over a decade ago sparked widespread enthusiasm for the development of new models of human disease.
  115. [115]
    Induced Pluripotent Stem Cells in Cardiovascular Drug Discovery
    Feb 1, 2013 · iPSC-based disease models represent a powerful new tool for drug discovery, with promising applications for primary phenotypic screening ...<|separator|>
  116. [116]
    Drug discovery using induced pluripotent stem cell models of ...
    The revolution of induced pluripotent stem cell (iPSC) technology provides a platform for development of cell therapy, disease modeling and drug discovery.
  117. [117]
    Neurological Disease Modeling Using Pluripotent and Multipotent ...
    Apr 21, 2023 · Neurological disease modeling using pluripotent and multipotent stem cells: a key step towards understanding and treating mucopolysaccharidoses.
  118. [118]
    An Overview of Pluripotent and Multipotent Stem Cell Targets
    Adult stem cells such as neural stem cells (NSCs), mesenchymal stem cells (MSCs), and hematopoietic stem cells (HSCs) are multipotent stem cells. The ...
  119. [119]
    Induced Pluripotent Stem Cells (iPSC) and Their Use in Disease ...
    Oct 7, 2025 · The development of iPSC technology by Shinya Yamanaka and colleagues in 2006 at Kyoto University represented a paradigm shift in ...
  120. [120]
    Harnessing the Potential of Stem Cells for Disease Modeling - NIH
    In this review, we discuss important breakthroughs of stem cell-based models highlighting current directions, advantages, and limitations.
  121. [121]
    Reprogramming barriers and enhancers: strategies to ... - NIH
    Nov 11, 2015 · The current review provides an in-depth overview of the cutting-edge findings regarding distinct barriers of reprogramming to pluripotency and strategies to ...
  122. [122]
    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 ...
  123. [123]
    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 ...
  124. [124]
    Genomic Instability of iPSCs: Challenges Towards Their Clinical ...
    Sep 5, 2016 · Here we summarize our current knowledge on genomic instability of iPSCs, with a particular focus on types of genetic variations and their origins.
  125. [125]
    Genomic integrity of human induced pluripotent stem cells across ...
    The genomic region of instability on chr20 (chr20q11.2) includes transcriptomic signatures for cancer associated genes such as ID1, BCL2L1, TPX2, PDRG1 and HCK.
  126. [126]
    Progress, Obstacles, and Limitations in the Use of Stem Cells ... - NIH
    One of the biggest obstacles to the use of stem cells is the variability in efficiency and robustness of differentiation protocols. The differentiation of stem ...2. Current Stem Cell... · 2.4. Stem Cell... · 3. Biological Tissue Sources...
  127. [127]
    Prevention of tumor risk associated with the reprogramming of ... - NIH
    This review focuses on the risk of tumor formation by human pluripotent stem cells, and on the possible treatment options if it occurs.
  128. [128]
    'Above all, do no harm': safeguarding pluripotent stem cell therapy ...
    Jun 3, 2014 · Every pluripotent undifferentiated stem cell poses a risk of neoplasmic transformation. Therefore, the aforementioned or other novel strategies ...
  129. [129]
    Reducing genomic instability in iPSCs - PMC
    Genomic instability in pluripotent cells has been detected since the early 2000's when karyotypically abnormal human ESC lines were observed following their ...
  130. [130]
    The Challenges to Advancing Induced Pluripotent Stem Cell ...
    Nov 29, 2023 · iPSC therapies pose significant unique challenges with respect to safety, potency, genetic stability, immunogenicity, tumorgenicity, cell reproducibility, ...
  131. [131]
    Improving the safety of human pluripotent stem cell therapies using ...
    Jun 1, 2020 · Here we use genome editing to engineer a general platform to improve the safety of future hPSC-derived cell transplantation therapies.
  132. [132]
    Examining the ethics of embryonic stem cell research
    Opponents argue that the research is unethical, because deriving the stem cells destroys the blastocyst, an unimplanted human embryo at the sixth to eighth day ...
  133. [133]
    Ethical Issues in Stem Cell Research - PMC - NIH
    Apr 14, 2009 · Several ethical concerns come into play when a frozen embryo is donated, including informed consent from the woman or couple donating the embryo ...
  134. [134]
    Ethical and Safety Issues of Stem Cell-Based Therapy - PMC - NIH
    The main safety issue regarding iPSC-based therapy (labeled with question marks) is the risk of teratoma formation which might happen if patient receive iPSC- ...
  135. [135]
    The Media Is Criticized For Embryonic Stem Cell Bias
    According to the Culture and Media Institute, members of the general media are inaccurately biased in favor of embryonic stem cell research, at the expense ...Missing: academia | Show results with:academia
  136. [136]
    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 ...
  137. [137]
    [PDF] FDA Perspective on the Ethics of Stem Cell Therapy
    “From a regulatory perspective, FDA considers stem cell- based products to be somatic cellular therapies that do not warrant a distinct regulatory approach.” • ...
  138. [138]
    Regulatory considerations for pluripotent stem cell therapies
    This review discusses current US cell therapy regulations and strategies for compliance with these regulations when developing PSC-derived products.
  139. [139]
    Pluripotent stem-cell-derived therapies in clinical trial: A 2025 update
    Jan 25, 2025 · Here, we review the landscape of interventional hPSC trials worldwide, highlighting available data on clinical safety and efficacy.
  140. [140]
    Comparative analysis of regulations and studies on stem cell therapies
    Nov 21, 2024 · In the United States, although there are no formal laws or regulations that restrict germline cell therapy, the FDA prohibits the use of ...
  141. [141]
    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.