Fact-checked by Grok 2 weeks ago

Cellular differentiation

Cellular differentiation is the by which a less specialized, immature progressively acquires the specialized structures, functions, and characteristics of a mature , enabling it to perform specific roles within a . This transformation typically begins with totipotent cells, such as the , which can develop into any including extra-embryonic tissues, and proceeds through stages of decreasing potency—pluripotent (capable of forming most body types), multipotent (restricted to specific lineages like blood cells), oligopotent (limited to a few related types), and unipotent (committed to one type)—ultimately resulting in approximately 400 distinct types, as estimated by recent studies (2023). The process is fundamental to embryonic development, where rapid cell divisions from a single fertilized egg generate the diverse and organs necessary for organismal form and function, and it persists throughout life to support growth, maintenance, and regeneration in response to injury or wear. In adult organisms, stem cells in niches such as or continually differentiate to replace lost or damaged cells, ensuring and longevity across species from to animals. Dysregulation of differentiation underlies numerous , including cancer—where cells revert to less differentiated states promoting uncontrolled —and degenerative conditions like or heart disease, where impaired differentiation hinders repair. At the molecular level, cellular differentiation is orchestrated by intricate regulatory mechanisms involving differential gene expression, where specific transcription factors bind to DNA to activate or repress genes, leading to the production of proteins that define cell identity and morphology. Extrinsic signals from the microenvironment, such as growth factors, hormones, and mechanical forces, interact with intrinsic genetic programs and epigenetic modifications—like DNA methylation and histone alterations—to guide lineage commitment and ensure precise, robust cell fate decisions. Recent advances, including the generation of induced pluripotent stem cells (iPSCs) through reprogramming with factors like Oct4, Sox2, Klf4, and c-Myc, have revealed the plasticity of differentiation, allowing differentiated cells to revert to a stem-like state for therapeutic applications.

Overview and Fundamentals

Definition and Process

Cellular differentiation is the process by which a less specialized becomes a more specialized , characterized by changes in that alter the cell's structure, function, and biochemistry without modifying the underlying sequence. This process is fundamental to the development of multicellular organisms, enabling the formation of diverse tissues and organs from a single fertilized . In essence, it involves the progressive restriction of a cell's developmental potential, transforming totipotent cells—such as the , which can give rise to all types including extraembryonic tissues—into cells with narrower fates. The process unfolds through distinct stages: specification, commitment (often termed determination), and realization (differentiation proper). Specification represents an initial, reversible assignment of fate, where a cell or group of cells can autonomously develop into a particular type if isolated in a neutral environment, but this commitment can still be altered by external cues. Commitment follows as an irreversible decision, where the cell's fate is fixed and will proceed even if placed in a different embryonic context, reflecting stable changes in its regulatory state. Finally, realization entails the overt morphological and functional changes that produce a mature, specialized cell, such as the extension of axons in neurons or the accumulation of hemoglobin in erythrocytes. In multicellular organisms, cellular differentiation culminates in the generation of terminally differentiated cells that perform specific roles, contributing to tissue and organ formation. For instance, during embryogenesis, the totipotent undergoes to form a blastula, a hollow ball of undifferentiated cells, which then reorganizes during into a gastrula with three primary germ layers: the (giving rise to and ), (forming muscle, , and ), and (producing the gut and associated organs). These layers arise through coordinated differentiation events, establishing the foundational . Stem cells, including pluripotent embryonic stem cells derived from the , serve as key starting points for subsequent differentiation pathways in both embryonic and adult contexts.

Historical Development

The foundations of cellular differentiation were laid in the through Rudolf Virchow's formulation of , which posited that all cells arise from pre-existing cells, emphasizing the cellular basis of development and disease. This work, detailed in his 1858 lectures compiled as Die Cellularpathologie, integrated with pathological processes, highlighting how cellular changes drive organismal differentiation. Building on this, August Weismann's theory in 1892 distinguished between germ cells, which transmit hereditary information, and cells, which undergo differentiation without altering the , thus separating reproductive continuity from somatic development. In the early , experimental advanced understanding through Hans Driesch's separation experiments on embryos in the 1890s, demonstrating regulative development where isolated blastomeres could form complete larvae, challenging mosaic theories and revealing cellular plasticity in early differentiation. Later, Conrad Waddington's epigenetic landscape model in the 1940s conceptualized differentiation as a ball rolling down branching valleys on an inclined surface, illustrating how genetic and environmental factors canalize cells toward specific fates without altering the genome. Mid-20th-century breakthroughs included John Gurdon's 1962 experiments in frogs, where cell nuclei reprogrammed in enucleated oocytes produced fertile adults, proving that differentiated nuclei retain totipotency and can reverse differentiation. Later, in the late , the discovery of master regulators like in 1987 showed how a single could convert fibroblasts to myoblasts, establishing key molecular switches for lineage commitment. This culminated in Shinya Yamanaka's identification of four s (Oct4, , , c-Myc) in 2006, enabling of cells into induced pluripotent cells, reviving totipotency and transforming views on cellular .

Cell Types and Differentiation Patterns

Stem Cells and Progenitors

Stem cells and progenitor cells represent the foundational of undifferentiated cells that drive cellular differentiation, characterized by their capacity to self-renew and generate specialized progeny. Totipotent cells exhibit the highest potency, exemplified by the , which can develop into all cell types of the organism, including extraembryonic tissues. Pluripotent stem cells, such as embryonic stem cells, possess the ability to differentiate into any cell type derived from the three primary germ layers (, , and ) but cannot form extraembryonic structures. This potency enables them to contribute to the formation of all bodily tissues during early embryonic development. Multipotent stem cells have a more restricted differentiation potential, capable of producing multiple, but lineage-specific, cell types; hematopoietic stem cells, for instance, reside in the and give rise to various lineages including erythrocytes, leukocytes, and platelets. Unipotent progenitors represent the narrowest scope, committed to generating a single within a lineage; spermatogonial stem cells in the testis serve as a prime example, solely producing spermatozoa to sustain . These progressive restrictions in potency reflect the hierarchical progression from broad embryonic potentials to specialized adult progenitors. A defining feature of s is their self-renewal capacity, maintained through symmetric cell divisions that produce two identical daughters or asymmetric divisions that yield one self-renewing and one destined for . This dual mechanism ensures population stability while allowing controlled expansion of , balancing with the onset of lineage commitment. , in turn, exhibit limited self-renewal and primarily undergo proliferative divisions to amplify cells prior to terminal . Stem cells are sourced from diverse origins to support developmental and regenerative needs. Embryonic stem cells are isolated from the of the blastocyst-stage , providing a renewable population for early formation. persist in specialized niches throughout life, such as the for hematopoietic lineages or the intestinal crypts where Lgr5-positive cells drive epithelial renewal. Induced pluripotent stem cells, generated in laboratories by adult cells through overexpression of key transcription factors like Oct4, , , and c-Myc, offer an ethical and patient-specific alternative with pluripotency akin to embryonic cells. In organismal development, stem cells and progenitors are essential for sustaining homeostasis and enabling regeneration after injury. They maintain steady-state renewal in high-turnover tissues, such as the and gut , by continuously replenishing differentiated cells. For example, neural stem cells in the adult and support limited , contributing to and repair. Through these roles, stem cells initiate differentiation hierarchies that ensure long-term integrity.

Terminal Differentiation and Cell Types

Terminal differentiation represents the final stage of cellular , during which cells irreversibly commit to specific functions and typically lose their capacity for , ensuring stable in mature organisms. This process results in cells that are highly adapted for particular roles, such as , , or , and is a hallmark of multicellular complexity in mammals. In mammals, terminally differentiated cells are broadly categorized into four primary tissue types: epithelial, , muscle, and , with blood cells often considered a specialized subset of connective tissue. Epithelial cells, for instance, include , which form a protective barrier by producing and undergoing to create the . Connective tissue features osteoblasts that secrete bone matrix components like and , eventually embedding themselves as non-proliferative osteocytes. Muscle cells encompass cardiomyocytes, which exhibit synchronized contractions for heart function through specialized structures. Nervous tissue is exemplified by neurons, which transmit electrical signals via extended axons and dendrites. Blood cells include erythrocytes, which mature by extruding their and synthesizing for oxygen transport. These categories arise from cells through lineage-specific pathways. Mammals possess over 200 distinct terminally differentiated cell types, each tailored to niche functions within organs. Notable examples include pancreatic beta cells, which produce and secrete insulin in response to glucose levels, maintaining metabolic balance. Hepatocytes in the liver perform , protein , and , supporting systemic . These specialized cells highlight the diversity of differentiation outcomes in mammalian . Functional adaptations in terminally differentiated cells often involve profound morphological and biochemical changes to optimize performance. For example, neurons extend axons up to a meter in length during to facilitate rapid signal propagation across the body. In erythrocytes, terminal maturation includes the accumulation of , enabling efficient oxygen binding and delivery while the loss of organelles streamlines circulation. Such adaptations underscore the precision of in achieving tissue-specific efficacy.

Core Mechanisms

Gene Regulatory Networks

Gene regulatory networks (GRNs) consist of interconnected circuits of transcription factors, enhancers, and feedback loops that precisely control the activation or repression of lineage-specific genes during cellular differentiation. These networks function as genomic logic systems, where transcription factors bind to cis-regulatory modules such as enhancers—clusters of DNA sequences typically exceeding 300 base pairs with multiple binding sites—to drive spatial and temporal gene expression patterns essential for developmental progression. Feedback loops within GRNs provide stability and robustness; for instance, positive feedback reinforces commitment to a differentiation state, while negative feedback sharpens boundaries between cell fates. Central to GRNs are master regulatory transcription factors that initiate and coordinate differentiation cascades by activating downstream targets. In , Pax6 serves as a master regulator, capable of inducing ectopic eye formation across diverse species; ectopic expression of the Drosophila homolog eyeless triggers the formation of functional compound eyes on appendages like wings, activating a battery of ~2500 eye-specification genes. Similarly, Gata4 acts as a master regulator in cardiac mesoderm differentiation, where its absence in knockout mice leads to defective ventral morphogenesis and failure of cardiac progenitor migration to the midline, despite initial specification of primitive myocytes expressing contractile proteins. These master regulators integrate inputs to orchestrate tissue-specific programs, ensuring fidelity in lineage commitment. GRNs exhibit dynamic architectures that govern the timing and rapidity of differentiation events. Feed-forward loops (FFLs), a prevalent in these networks, enable rapid of genes while filtering ; in a coherent type-1 FFL, an upstream activator regulates both a direct and an intermediate , allowing swift responses to sustained signals during cell fate transitions. Oscillatory networks, such as those involving Hes/Her genes, drive periodic processes like somitogenesis through the segmentation clock; Hes7, a transcriptional , oscillates via delayed on its own expression, with a period of ~2 hours in mice, coordinating sequential formation essential for vertebral patterning. These dynamics ensure synchronized differentiation across cell populations. A prominent example of GRN logic in differentiation is the hematopoietic cascade, where antagonistic interactions between transcription factors dictate myeloid versus erythroid lineages from hematopoietic stem cells. High levels of Pu.1 promote myeloid differentiation by directly inhibiting Gata1 through protein-protein interaction and competition for DNA binding sites on shared target promoters, blocking erythroid gene activation; conversely, elevated Gata1 represses Pu.1, reinforcing erythroid commitment and terminal maturation. This mutual antagonism forms a bistable switch within the GRN, enabling robust lineage bifurcation. GRNs integrate signaling inputs, such as those from cytokines, to trigger activation thresholds in these circuits.

Cell Signaling Pathways

Cell signaling pathways play a crucial role in cellular differentiation by transducing extracellular cues from the microenvironment into intracellular responses that direct fate decisions. These pathways typically begin with the of soluble ligands, such as factors or morphogens, to specific surface receptors, initiating cascades that propagate signals through the to modulate in the . This process ensures precise spatiotemporal control during embryogenesis and tissue homeostasis, where signals integrate to promote or inhibit differentiation toward specific lineages. Among the major pathways, the Wnt/β-catenin pathway is essential for establishing and fate in early development, such as anterior-posterior axis formation in vertebrates. In this canonical pathway, Wnt ligands bind to receptors and LRP5/6 co-receptors, leading to the stabilization and nuclear translocation of β-catenin, which then activates transcription factors like TCF/LEF to drive target favoring over . For instance, in embryonic stem cells, Wnt signaling promotes mesodermal by repressing pluripotency genes.01344-4) The pathway mediates direct cell-cell communication, particularly through , which refines patterns during and somitogenesis. receptors on the signal-receiving cell are activated by transmembrane ligands like or on adjacent cells, triggering sequential proteolytic cleavages that release the intracellular domain (NICD). NICD translocates to the and forms a complex with RBPJ to induce transcription of hes/her genes, suppressing neuronal in progenitors while allowing it in neighbors. This binary signaling ensures balanced production of differentiated cell types in tissues like the . Hedgehog (Hh) signaling governs patterning and differentiation in structures like limbs and the , acting as a to specify positional identity. In the absence of Hh ligands (e.g., Sonic ), the receptor Patched inhibits (Smo); ligand binding relieves this inhibition, activating Smo to prevent Gli degradation and promote its nuclear activity for target gene expression. This pathway is critical for ventral differentiation, where graded Hh activity induces distinct neuronal subtypes. The TGF-β superfamily, including , regulates induction and dorsal-ventral patterning through serine/ receptors that phosphorylate Smad proteins. Upon binding, receptor-activated Smads (R-Smads) complex with Smad4 and enter the to regulate transcription, often promoting in a concentration-dependent manner. For example, gradients in the early establish dorsal-ventral axes by inducing ventral at high levels and neural tissue dorsally via inhibition. (FGF) signaling complements this in limb development, where FGFs from the apical ectodermal ridge sustain outgrowth and proximodistal patterning by activating MAPK/ERK cascades that maintain undifferentiated progenitors at the bud tip while allowing proximally. These pathways exhibit extensive crosstalk to fine-tune outcomes; for instance, Wnt signaling can antagonize activity by inducing antagonists like , preventing excessive mesodermal specification during . Mechanisms of often involve ligand-receptor interactions without classical second messengers like , though G-protein-coupled receptors in some contexts (e.g., non-canonical Wnt) utilize to modulate downstream effectors. Overall, these cascades converge on regulatory networks to orchestrate programs.

Inductive and Asymmetric Division

Inductive signaling refers to the process by which one group of cells influences the developmental fate of neighboring cells through secreted or contact-dependent signals, thereby propagating cues across tissues. A classic example is the Spemann-Mangold organizer in embryos, where the dorsal blastopore lip induces the overlying to form the , demonstrating how localized signaling centers can direct spatial patterning during . This interaction ensures that undifferentiated cells adopt specific fates in response to positional information from inducing tissues. Asymmetric cell division, in contrast, generates cellular diversity within a by unequally partitioning cytoplasmic determinants, organelles, or signaling components between daughter cells, often resulting in one maintaining stem-like properties while the other differentiates. In the Caenorhabditis elegans, divisions exemplify this, where polarity cues lead to one daughter retaining self-renewal capacity and the other committing to a differentiated neuronal fate, though the specific determinant Numb is more prominently featured in analogous processes in other organisms.90112-0) Such divisions are crucial for maintaining pools while producing differentiated progeny in a controlled manner. Key mechanisms underlying these processes include the establishment of cellular polarity through PAR proteins, which form opposing cortical domains to direct the asymmetric localization of fate determinants during . In C. elegans, PAR-3 and PAR-6 localize to the anterior , while PAR-1 and PAR-2 enrich posteriorly, creating a scaffold that biases spindle orientation and determinant segregation. Contact-dependent signaling further refines outcomes, as seen in the - pathway during the asymmetric division of sensory organ precursor (SOP) cells, where the ligand Delta on one daughter activates Notch in the sibling, promoting differential fates without requiring extrinsic inducers.90112-0) Representative examples illustrate these principles in diverse contexts. In mammalian intestinal crypts, Lgr5-positive s undergo asymmetric divisions influenced by apicobasal polarity and Numb segregation, yielding one renewing and one transit-amplifying progenitor that differentiates into absorptive or secretory lineages. Similarly, vulval in C. elegans involves the anchor secreting LIN-3 (an EGF-like signal) to induce adjacent vulval precursor s to adopt primary, secondary, or tertiary fates, highlighting how inductive interactions pattern epithelial structures through graded signaling. These mechanisms collectively ensure the spatiotemporal coordination of , balancing maintenance and specialization.

Epigenetic Regulation

Importance in Differentiation

Epigenetics refers to mitotically heritable changes in gene activity that occur without alterations to the underlying sequence. These modifications, including and histone alterations, enable cells to maintain distinct identities across cell divisions, ensuring that patterns are stably propagated in daughter cells. In cellular differentiation, epigenetic mechanisms play a pivotal role in locking in differentiated states after initial signaling cues have directed fate decisions, thereby preventing reversion to more pluripotent or undifferentiated conditions. For instance, during the transition from pluripotent cells to lineages, epigenetic silencing represses pluripotency-associated genes such as Oct4 and Nanog, establishing a stable environment that enforces commitment and restricts cellular plasticity. This heritable stability is initiated by transient signaling pathways that trigger epigenetic , but the modifications themselves provide the enduring framework for identity.01446-7) Disruption of these epigenetic controls can lead to aberrant , prominently observed in diseases like cancer where loss of repressive marks such as promotes and reactivation of developmental pathways.30358-1) In malignancies, diminished levels at promoter regions allow inappropriate expression of lineage-inappropriate genes, contributing to tumor heterogeneity and aggressive phenotypes. Evidence for the critical role of in comes from studies of identical twins, which reveal progressive epigenetic divergence despite shared genomes, highlighting how environmental and developmental cues drive heritable changes that influence cell fate. In aging twin pairs, epigenetic profiles become markedly distinct, with variations in patterns correlating to differences in and phenotypic outcomes. Similarly, genetic knockout models demonstrate these effects; for example, Dnmt1-deficient mice exhibit impaired erythroid due to disrupted maintenance of , leading to arrest in erythroid progenitors and failure to progress to mature red blood cells.

Specific Epigenetic Mechanisms

Histone modifications serve as key epigenetic regulators during cellular by altering structure and accessibility to influence . Activating marks, such as trimethylation of at 4 (H3K4me3), are deposited by Trithorax group (TrxG) complexes and are enriched at promoters of lineage-specific genes to promote their in differentiating cells. For instance, in embryonic stem cells (ESCs) transitioning to lineage-committed states, H3K4me3 marks facilitate the expression of developmental genes by maintaining open configurations. In contrast, repressive modifications like trimethylation of at 27 (H3K27me3), catalyzed by Polycomb repressive complex 2 (PRC2), silence pluripotency-associated genes during . A prominent example occurs in the ESC-to-neural progenitor transition, where PRC2-mediated H3K27me3 represses pluripotency factors such as Oct4, enabling neurogenic gene .00187-7) These bivalent domains, characterized by co-occurrence of H3K4me3 and H3K27me3 at developmental loci in ESCs, resolve upon to favor either or stable repression.00187-7) DNA methylation, primarily involving the addition of a to the 5' position of (5-, 5mC) at CpG islands, acts as a repressive epigenetic mark that stabilizes in differentiated cells. During , de novo DNA methylation by DNA methyltransferases (DNMTs) targets promoters of pluripotency genes, such as the Oct4 distal enhancer and proximal promoter, leading to their transcriptional repression and commitment to specific lineages. This hypermethylation prevents reactivation of pluripotency networks, ensuring terminal states. In reprogramming contexts, active demethylation is facilitated by ten-eleven translocation (TET) enzymes, which oxidize 5mC to 5- (5hmC) and subsequent intermediates, promoting locus-specific erasure of methylation to restore pluripotency . TET-mediated demethylation is essential for efficient induced pluripotency, as its disruption impairs the activation of key reprogramming targets.00002-2) Chromatin remodeling complexes dynamically reposition to modulate DNA accessibility during . The family of ATP-dependent remodelers, including BAF and PBAF variants, slides, ejects, or exchanges to expose or conceal regulatory elements, thereby directing lineage-specific gene programs. In ESCs, complexes maintain pluripotency by repressing genes, but upon cues, they facilitate access to lineage enhancers. transcription factors, such as Oct4, , and Nanog, bind directly to closed regions, initiating nucleosome displacement and opening to recruit additional factors. For example, Oct4 cooperates with the BRG1 subunit of to enhance accessibility at pluripotency enhancers, supporting both self-renewal and the initial stages of . This activity establishes competence for subsequent regulatory events without requiring prior opening. Other epigenetic mechanisms, including non-coding RNAs and histone variants, further refine differentiation outcomes. The long non-coding RNA coats the inactive in female cells, recruiting silencing complexes to achieve dosage compensation through , a differentiation-dependent process essential for proper embryonic development. variants like H2A.Z, incorporated at promoter and enhancer regions, modulate stability and facilitate the binding of both activating and repressive complexes. In ESCs, H2A.Z enrichment at bivalent promoters poises genes for activation during differentiation, while its depletion impairs both self-renewal and lineage commitment by altering chromatin accessibility. These mechanisms collectively ensure stable, heritable changes in patterns that define cellular identity.

Integration with Signaling and Environment

Cellular differentiation is profoundly influenced by the dynamic interplay between external signaling cues and epigenetic modifications, where signaling pathways directly recruit or modulate epigenetic regulators to alter chromatin states and gene expression. In the Wnt/β-catenin pathway, stabilization of β-catenin upon ligand binding leads to its nuclear translocation, where it binds to promoters and displaces the Polycomb Repressive Complex 2 (PRC2), specifically its catalytic subunit EZH2, thereby relieving repression and allowing activation of target genes essential for stem cell maintenance and differentiation during embryonic development. Similarly, activation of the Notch pathway releases the Notch intracellular domain (NICD), which translocates to the nucleus and recruits histone acetyltransferases (HATs) such as p300 and PCAF to Notch target gene enhancers, promoting histone acetylation and transcriptional activation critical for cell fate decisions in neurogenesis and other lineages. This crosstalk ensures that transient signals translate into stable epigenetic changes, such as alterations in core marks like H3K27me3, to commit cells to specific differentiation trajectories. Environmental physical cues, particularly (ECM) stiffness, integrate with epigenetic regulation to guide lineage specification through mechanotransduction pathways. On soft substrates mimicking neural tissues (approximately 0.1–1 kPa), mesenchymal stem cells (MSCs) exhibit nuclear exclusion of the transcriptional coactivators /TAZ, which prevents their binding to promoters of neurogenic repressors and instead favors epigenetic activation of neuronal genes, such as those involving the neuron-restrictive silencer factor (NRSF), leading to enhanced . Conversely, stiff matrices (around 25–40 kPa) emulate environments and promote nuclear localization of /TAZ, which cooperates with to drive histone modifications supporting osteogenic , including increased at RUNX2 target loci and elevated levels of osteogenic markers like . These mechanosensitive interactions highlight how biomechanics fine-tune epigenetic landscapes to direct differentiation without relying solely on soluble factors. Metabolic signals, such as nutrient availability, further modulate epigenetic states during by providing substrates for chromatin-modifying enzymes. , a key methyl donor in the one-carbon pathway, is essential for during neural tube closure; its deficiency impairs global patterns, particularly at imprinted genes and developmental regulators, increasing the risk of neural tube defects (NTDs) by disrupting epigenetic silencing required for proper neural progenitor specification. Supplementation restores methylation fidelity, underscoring the nutrient's role in linking to epigenetic stability in embryogenesis. In stem cell niches, hypoxia-inducible factors (HIFs) exemplify how microenvironmental oxygen levels interface with to regulate . Low oxygen stabilizes HIF-1α, which enhances H3K9 at pluripotency genes in human embryonic stem cells, maintaining an open state that supports self-renewal while priming commitment upon reoxygenation; this is mediated through recruitment of HAT complexes to HIF-responsive enhancers. In hematopoietic and neural niches, HIFs similarly alter to balance quiescence and , ensuring niche-specific epigenetic adaptations to hypoxic stress.

Dedifferentiation and Reprogramming

Natural Dedifferentiation

Natural dedifferentiation is the process by which differentiated cells spontaneously revert to a less specialized state, losing mature phenotypic traits while acquiring proliferative capacity or stem-like potential in response to physiological cues such as or regeneration demands. This reversal contrasts with the typical unidirectional progression of during and enables tissue repair in certain organisms without relying on dedicated pools. In this context, dedifferentiation facilitates the reactivation of embryonic-like programs, allowing cells to contribute to new tissue formation while maintaining fidelity. Prominent examples of natural dedifferentiation occur in amphibian regeneration. In newts, lens regeneration initiates when dorsal iris pigment epithelial cells dedifferentiate following lentectomy, depigmenting and proliferating to form a new lens vesicle through transdifferentiation. This process is asymmetric, involving only dorsal iris cells and highlighting intrinsic cellular plasticity unique to urodeles. Similarly, in salamander limb regeneration, differentiated skeletal myofibers dedifferentiate by fragmenting into mononucleate cells that reenter the cell cycle, retaining early myogenic markers like Pax7 while downregulating terminal differentiation genes such as myosin heavy chain. These dedifferentiated cells migrate to form the blastema, a proliferative mound that rebuilds the entire limb structure. Mechanistically, natural involves transient epigenetic remodeling to erase differentiation-associated marks and unlock proliferative genes. For instance, in limb regeneration, changes in dynamics, including upregulation of DNMT3a, accompany formation and correlate with the upregulation of pluripotency factors, facilitating the transition from quiescent myofibers to proliferative progenitors. Injury-response genes, such as Lin28, are rapidly activated to support this shift; Lin28 enhances by promoting metabolic reprogramming toward , boosting , and suppressing let-7 microRNAs that enforce . In salamander models, Lin28 expression surges post-amputation, coordinating with pathways like Wnt and to sustain the dedifferentiated state temporarily before redifferentiation. In mammals, natural dedifferentiation is more restricted, often limited to partial reversals in response to injury rather than full regenerative capacity. During skin wound healing, terminally differentiated Gata6+ from follicles dedifferentiate, re-expressing markers like Itga6 and Lrig1 to migrate, proliferate, and repopulate the , driven by activation and signaling. In the heart, post-injury cardiac fibroblasts exhibit enhanced plasticity through activation and differentiation into myofibroblasts, increasing proliferation and remodeling, though this typically leads to rather than regeneration. These mammalian instances underscore the evolutionary of dedifferentiation, relying on endogenous signals like but lacking the robust epigenetic erasure seen in amphibians.

Induced Pluripotent Stem Cells

Induced pluripotent stem cells (iPSCs) are generated by reprogramming cells to a pluripotent state resembling embryonic stem cells, enabling them to differentiate into any cell type of the three germ layers. This technique was pioneered in 2006 when Kazutoshi Takahashi and demonstrated that mouse embryonic and adult fibroblasts could be reprogrammed to pluripotency by introducing four transcription factors: Oct4, , , and c-Myc, collectively known as the Yamanaka factors. These factors activate pluripotency genes while suppressing identities, marking a breakthrough that built on prior experiments demonstrating cellular plasticity. The method was extended to human cells in 2007 using the same factors on dermal fibroblasts, confirming its applicability across species. Various methods have been developed to introduce these reprogramming factors, evolving from initial integrative approaches to safer, non-integrative ones to avoid genomic disruptions. Early protocols relied on viral vectors, such as retroviruses or lentiviruses, which integrate into the host to express the factors, achieving high efficiency but risking . Non-integrating alternatives include Sendai virus, a that self-eliminates after , reducing oncogenic risks while maintaining comparable efficiency. Other non-integrative techniques involve synthetic mRNA delivery, which transiently expresses factors without genomic integration, or episomal plasmids that persist temporarily in the . Additionally, small molecules can replace or augment transcription factors; for instance, inhibitors of Polycomb Repressive Complex 2 (PRC2), such as inhibitors, facilitate epigenetic reset by alleviating H3K27me3-mediated repression of pluripotency loci, enhancing reprogramming efficiency in human cells. The reprogramming process involves a stochastic, multi-step epigenetic reconfiguration that erases memory and establishes a pluripotent epigenome. Initially, the Yamanaka factors initiate mesenchymal-to-epithelial transition and partial activation of pluripotency networks, accompanied by global opening and at loci like Oct4 and Nanog, which reactivates endogenous pluripotency genes. This is followed by progressive silencing of genes through mechanisms such as de novo and histone modifications, including increased at lineage-specific enhancers via PRC2 activity. Over time, the epigenome undergoes metastable changes, with intermediate states marked by bivalent domains (co-occupancy of activating and repressive ) that poise genes for ; full pluripotency is achieved when these resolve into an open, accessible landscape similar to embryonic cells. The process typically takes 2-4 weeks, with efficiency around 0.1-1% for standard methods, though optimizations can improve yields. iPSCs have transformative applications in disease modeling and due to their ability to derive patient-specific cells for personalized studies and therapies. In disease modeling, patient-derived iPSCs harboring genetic mutations enable the generation of relevant cell types to recapitulate pathologies; for example, iPSCs from patients differentiate into neurons exhibiting α-synuclein aggregation and mitochondrial dysfunction, facilitating drug screening and mechanistic insights. This approach has advanced understanding of sporadic and familial forms, with models revealing impaired and as key features. In , iPSC-derived cells are tested in clinical trials; notably, autologous iPSC-derived retinal pigment epithelial cells have been transplanted into patients with age-related , demonstrating safety and modest visual improvements in phase I trials without tumorigenicity. Similarly, allogeneic iPSC-derived progenitors are in phase I/II trials for as of 2025, showing survival, dopamine production, and no tumors, aiming to restore motor function by engrafting into the . These applications underscore iPSCs' potential to address unmet needs in neurodegenerative and degenerative diseases, though challenges like epigenetic memory and scalability persist.

Evolutionary Aspects

Origins and Early Evolution

The earliest manifestations of cellular differentiation arose in prokaryotes through functional division of labor among cells, predating multicellularity. In filamentous , specialized heterocysts evolved to perform , a incompatible with the oxygenic of vegetative cells. These thick-walled cells protect the oxygen-sensitive enzyme by repressing and forming a barrier, allowing atmospheric N₂ conversion to for the filament. Phylogenetic and evidence indicates this innovation emerged between 2.45 and 2.1 billion years ago, contemporaneous with the that increased atmospheric oxygen levels. In unicellular eukaryotes, rudimentary differentiation-like states provided precursors to more complex cellular specialization. yeasts exhibit , a reversible epigenetic process where haploid cells alter their (a or α) via cassette recombination, enabling and diploid formation. This mechanism has independently evolved at least 11 times in lineages, highlighting its adaptive value in promoting without permanent cell fate commitment.30783-3) Similarly, in the amoebozoan , unicellular amoebae enter dormant encysted states under adverse conditions or aggregate during starvation to form transient multicellular fruiting bodies with stalk and cells. Dictyostelid multicellularity, diverging around 0.52 billion years ago, likely co-opted ancestral encystation pathways involving signaling for sporulation, bridging solitary and social lifestyles. The full transition to multicellularity, enabling stable , occurred approximately 1 billion years ago in eukaryotic lineages, as evidenced by colonial forms in choanoflagellates and volvocine algae. Choanoflagellates, the closest living relatives to metazoans, form adhesive rosette colonies via incomplete and bacterial-induced , recapitulating early steps in tissue formation without true germ-soma separation.01095-X)30769-4) In parallel, volvocine green algae like exhibit progressive complexity, with species such as Gonium forming small colonies and displaying complete germ-soma : somatic cells specialize in and uptake, while gonidia dedicate to . This separation evolved through trade-offs in cell size and function, enhancing colony fitness in larger groups. Critical innovations underpinning these transitions included cell adhesion molecules and signaling pathways. Cadherins, calcium-dependent transmembrane proteins mediating homophilic , originated before metazoans, with homologs identified in the pre-metazoan holozoan Capsaspora owczarzaki, facilitating colonial stability and morphogenesis in early animals. Concurrently, the , conserved across metazoans, emerged in an ancestral bilaterian or pre-metazoan context to establish anterior-posterior polarity and drive posterior differentiation, integrating environmental cues with fate decisions.30586-X) These molecular toolkits laid the foundation for hierarchical organization in multicellular life.

Conservation Across Species

Cellular differentiation exhibits remarkable genetic conservation across species, particularly through homologous transcription factors that orchestrate patterning and cell fate decisions. Hox genes, encoding homeodomain-containing transcription factors, are highly conserved among bilaterian animals and play a central role in anterior-posterior body axis patterning, which directs the differentiation of diverse cell types during embryogenesis. For instance, Hox clusters maintain collinear expression patterns from insects to vertebrates, ensuring spatial organization of differentiated tissues such as the vertebrate spinal cord. Similarly, the Pax6 transcription factor demonstrates profound conservation in eye development, functioning as a master regulator from Drosophila to humans; its paired and homeodomain motifs show over 90% sequence identity across these taxa, enabling Pax6 to induce ectopic eye structures when expressed in heterologous species like flies. This functional interchangeability underscores Pax6's role in specifying neuroectodermal progenitors and differentiating photoreceptor cells across metazoans. Epigenetic mechanisms regulating differentiation also display broad conservation, though with notable variations. Polycomb repressive complex 2 (PRC2), which deposits repressive marks to silence developmental genes, traces its core components (, EED, SUZ12, RBBP4/7) to the last eukaryotic common ancestor and is present in animals, , and fungi, where it maintains cellular identity during . Trithorax group (TrxG) complexes, counteracting PRC2 by promoting H3K4 methylation and active transcription, are similarly conserved across these kingdoms, ensuring stable activation of lineage-specific genes in processes like stem cell . , another key epigenetic layer, facilitates gene repression in vertebrates and by modifying residues in CpG contexts to stabilize differentiated states, such as in mammalian neural lineages or plant ; however, it is largely absent in , which relies more on modifications for equivalent functions.00190-0)00071-3) Despite these conserved elements, variations in differentiation plasticity highlight evolutionary adaptations, particularly in regeneration. Planarians exhibit high cellular plasticity through totipotent neoblasts, which proliferate and differentiate into all cell types post-injury, contrasting with mammals' reliance on lineage-restricted progenitors like cells, limiting regenerative scope to specific tissues such as muscle. In colonial like , budding blurs - boundaries, allowing multipotent cells to generate , an evolutionary innovation reflecting early flexibility in cell fate separation compared to vertebrates' strict early sequestration. From an evolutionary viewpoint, core differentiation pathways like Notch signaling have been co-opted from ancient metazoan origins, where it mediates to diversify cell fates in tissues from fly wings to human , balancing proliferation and . Such conservation comes with trade-offs in complex organisms: terminal , which locks cells into specialized, post-mitotic states for efficient tissue function, restricts regeneration, as seen in mammals where epigenetic barriers prevent , unlike in simpler regenerators like planarians. This evolutionary compromise enhances organismal complexity but curtails plasticity, potentially linking to reduced regenerative capacity in vertebrates.

References

  1. [1]
    Definition of cell differentiation - NCI Dictionary of Cancer Terms
    The process during which young, immature (unspecialized) cells take on individual characteristics and reach their mature (specialized) form and function.Missing: key aspects
  2. [2]
    Cellular Differentiation – Anatomy & Physiology - UH Pressbooks
    Differentiation is the process by which unspecialized cells become specialized to carry out distinct functions.Missing: definition | Show results with:definition
  3. [3]
    Key Definitions | Sue & Bill Gross Stem Cell Research Center
    Differentiation—The process whereby an unspecialized embryonic cell acquires the features of a specialized cell such as a heart, liver, or muscle cell.<|control11|><|separator|>
  4. [4]
    Rethinking differentiation: Stem cells, regeneration, and plasticity - NIH
    Cell differentiation is an essential process for the development, growth, reproduction and longevity of all multicellular organisms.
  5. [5]
    [PDF] Mechanical Control of Cell Differentiation: Insights from the Early ...
    Apr 6, 2022 · Differentiation is the process by which a cell activates the expression of tissue-specific genes, downregulates the expression of potency ...
  6. [6]
    The Developmental Mechanics of Cell Specification - NCBI - NIH
    Cell specification is the first stage of commitment where a cell can differentiate autonomously in a neutral environment, but this commitment can be reversed.Missing: realization | Show results with:realization
  7. [7]
    Totipotency: What It Is and What It Is Not - PMC - NIH
    Totipotent zygotes are distinct from pluripotent stem cells or tumors because they can originate development.
  8. [8]
    The Circle of Life: The Stages of Animal Development - NCBI
    As a result of gastrulation, the embryo contains three germ layers: the ectoderm, the endoderm, and the mesoderm. 3. Once the three germ layers are ...
  9. [9]
    Virchow's Contribution to the Understanding of Thrombosis and ...
    In 1858, Virchow presented a series of 20 lectures to a group of practicing physicians in Berlin at the Institute of Pathology. These lectures were ...
  10. [10]
    Das Keimplasma; eine Theorie der Vererbung - Internet Archive
    Feb 11, 2015 · Das Keimplasma; eine Theorie der Vererbung. by: Weismann, August, 1834-1914. Publication date: 1892 ... PDF download · download 1 file · SINGLE ...
  11. [11]
    "The Potency of the First Two Cleavage Cells in Echinoderm ...
    Jan 1, 2012 · The potency of the first two cleavage cells in echinoderm development. Experimental production of partial and double formations (1891-1892), by Hans Driesch.Missing: regulative | Show results with:regulative
  12. [12]
    Conrad Waddington and the origin of epigenetics
    Mar 15, 2015 · Denis Noble discusses Conrad Waddington's classic paper, 'The genetic assimilation of the bithorax phenotype', published in Evolution in 1956.
  13. [13]
    A glossary for stem-cell biology - Nature
    Jun 28, 2006 · Totipotent Sufficient to form entire organism. Totipotency is seen in zygote and plant meristem cells; not demonstrated for any vertebrate stem ...
  14. [14]
    Basics of Stem Cell Biology as Applied to the Brain - NCBI - NIH
    Jul 27, 2016 · In a way, SCNT is simply a special case of cell fusion of an enucleated totipotent zygote with a differentiated cell. The egg cell then goes on ...
  15. [15]
    Asymmetric and symmetric stem-cell divisions in development and ...
    Jun 29, 2006 · Asymmetric cell division is a defining characteristic of stem cells that enables them to simultaneously perpetuate themselves (self-renew) and generate ...
  16. [16]
    Stem Cell Basics
    Previous work with mouse embryos led to the development of a method in 1998 to derive stem cells from the inner cell mass of preimplantation human embryos and ...
  17. [17]
    Searching for adult stem cells in the intestine - PMC - PubMed Central
    Although there has been agreement that crypts contain 4–6 independent stem cells, two hypotheses exist regarding their exact identity and location.
  18. [18]
    Induced pluripotent stem cells (iPSCs): molecular mechanisms of ...
    Apr 26, 2024 · In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling ...
  19. [19]
    Integrating physiological regulation with stem cell and tissue ...
    This allows stem cells to sustain homeostasis or to remodel relevant tissues in response to physiological change. Stem cells are therefore not only regulated by ...
  20. [20]
    Adult neural stem cells in the mammalian central nervous system
    May 12, 2009 · Neural stem cells rescue nervous purkinje neurons by restoring molecular homeostasis of tissue plasminogen activator and downstream targets.
  21. [21]
    Cell Cycle Reactivation in Skeletal Muscle and Other Terminally ...
    According to a classic concept, a terminally differentiated (TD) cell is defined as one that, in the course of acquiring specialized functions, has irreversibly ...
  22. [22]
    Animal Cell Differentiation Patterns Suppress Somatic Evolution - PMC
    The series includes stem cells (stage 0, in white), TACs (in gray), and finally, terminally differentiated cells (stage 3, in black). Stem cells divide ...
  23. [23]
    Histology, Cell - StatPearls - NCBI Bookshelf - NIH
    Mar 27, 2025 · Cells are of different types, each with a unique function. Estimates suggest that the adult human body contains over 200 different cell types.
  24. [24]
    Old, new and emerging functions of caspases | Cell Death ... - Nature
    Dec 19, 2014 · Other cells that demonstrate enucletion during differentiation include RBCs and keratinocytes. Several caspases, including caspase-3, -2, and -9 ...
  25. [25]
    Skeletal muscle differentiation of human iPSCs meets ... - Nature
    Apr 7, 2022 · This review highlights the potential of this combination aimed at pushing the boundaries of skeletal muscle tissue engineering.Missing: categories | Show results with:categories
  26. [26]
    Axonal mRNA translation in neurological disorders - PMC
    It is increasingly recognized that local protein synthesis (LPS) contributes to fundamental aspects of axon biology, in both developing and mature neurons.Missing: erythrocytes | Show results with:erythrocytes
  27. [27]
    Cell Proliferation in Development and Differentiation - NCBI - NIH
    Early development is characterized by the rapid proliferation of embryonic cells, which then differentiate to produce the many specialized types of cells.Missing: definition | Show results with:definition
  28. [28]
    Mesenchymal stem cells lineage and their role in disease ...
    Nov 11, 2024 · Differentiated hepatocyte-like cells express several hepatocyte-specific markers and exhibit several biological functions in mature hepatocytes ...
  29. [29]
    The Erythroid Phenotype of EKLF-Null Mice: Defects in Hemoglobin ...
    Development of red blood cells requires the correct regulation of cellular processes including changes in cell morphology, globin expression and heme synthesis.
  30. [30]
    Gene regulatory networks for development - PNAS
    Mar 23, 2005 · The genomic program for development operates primarily by the regulated expression of genes encoding transcription factors and components of ...Gene Regulatory Networks For... · Sign Up For Pnas Alerts · Dorsal--Ventral Patterning...
  31. [31]
  32. [32]
    GATA4 transcription factor is required for ventral morphogenesis ...
    Analysis of cardiac development in the GATA4-/- mice demonstrated that these embryos developed splanchnic mesoderm, which differentiated into primitive cardiac ...
  33. [33]
    Structure and function of the feed-forward loop network motif - PNAS
    The FFL, a three-gene pattern, is composed of two input transcription factors, one of which regulates the other, both jointly regulating a target gene.
  34. [34]
    Signaling in Cell Differentiation and Morphogenesis - PubMed Central
    In this article, I review some of the signaling pathways that play essential roles during embryonic development.
  35. [35]
    Wnt/β-catenin signalling: function, biological mechanisms ... - Nature
    Jan 3, 2022 · The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis.
  36. [36]
    Notch signaling pathway: architecture, disease, and therapeutics
    Mar 24, 2022 · NOTCH signaling can rapidly regulate the dynamic transformation of cells to maintain physiological homeostasis, such as stem cells and tail ...
  37. [37]
    Concise Review: Notch Signaling in Stem Cell Systems
    Jan 2, 2009 · This report reviews the current knowledge of the Notch signaling pathway in various types of stem and early progenitor cells, but not germ cells.
  38. [38]
    The Hedgehog pathway: role in cell differentiation, polarity and ...
    The Hedgehog pathway regulates cell differentiation, proliferation, tissue polarity, stem cell population, and guides cell development as a morphogen.
  39. [39]
    A highlight on Sonic hedgehog pathway
    Mar 20, 2018 · Hedgehog (Hh) signaling pathway plays an essential role during vertebrate embryonic development and tumorigenesis.
  40. [40]
    The roles and regulatory mechanisms of TGF-β and BMP signaling ...
    Jan 24, 2024 · BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner.
  41. [41]
    Signaling cross-talk between TGF-β/BMP and other pathways - PMC
    Simple as it is, the TGF-β/BMP pathway controls a myriad of events, including cell proliferation, differentiation, apoptosis, migration, extracellular matrix ( ...
  42. [42]
    The BMP signaling gradient is interpreted through concentration ...
    Jan 22, 2021 · The BMP signaling gradient is interpreted through concentration thresholds in dorsal–ventral axial patterning · Identification of target genes ...
  43. [43]
    Limb Development - FGF Signalling in Vertebrate Development - NCBI
    Limb development is driven by the AER and ZPA, with FGF signaling mediating AER activity. FGF8 is sufficient to drive PD development.
  44. [44]
    Crosstalk between Wnt and bone morphogenetic protein signaling ...
    This review explores the molecular processes and mechanisms of the crosstalk between the Wnt and BMP signaling pathways in osteogenic differentiation and bone ...
  45. [45]
    Crosstalk between Wnt and bone morphogenic protein signaling: A ...
    Jun 19, 2009 · In those cases, while causing differentiation, BMP signaling blocks the effect of Wnt signals. This would prevent cells from receiving two ...INTRODUCTION · SYNERGISTIC AND... · MOLECULAR INTERACTIONS...
  46. [46]
    Spemann-Mangold Organizer | Embryo Project Encyclopedia
    Jan 12, 2012 · To explore neural plate induction, Spemann first performed a transplant experiment that was nearly identical to the later organizer experiment.Missing: seminal | Show results with:seminal
  47. [47]
    The Ins(ide) and Outs(ide) of Asymmetric Stem Cell Division - PMC
    In this review, we summarize critical aspects of asymmetric cell division with a particular focus on these and other emerging mechanisms that reinforce and ...
  48. [48]
    The gene lin-3 encodes an inductive signal for vulval development ...
    Aug 6, 1992 · The lin-3 gene is necessary for induction of the Caenorhabditis elegans vulva by the anchor cell. It encodes a molecule similar to epidermal growth factor.Missing: signaling seminal paper
  49. [49]
    Epigenetics and MicroRNAs | Pediatric Research - Nature
    Epigenetics is defined as mitotically and meiotically heritable changes in gene expression that do not involve a change in the DNA sequence.
  50. [50]
    DNA methylation: an epigenetic mark of cellular memory - Nature
    Apr 28, 2017 · Currently, the widely accepted definition of 'epigenetics' is 'heritable changes in genome function that occur without changes in the DNA ...
  51. [51]
    Epigenetic Mechanisms that Regulate Cell Identity - ScienceDirect
    Nov 5, 2010 · This mechanism is responsible for both the loss of cell plasticity during differentiation and the preservation of cell identity.
  52. [52]
    Epigenetic regulation of pluripotency and differentiation - PMC
    Here, we review the epigenetic control of pluripotency and changes associated with PSC differentiation. We focus on DNA methylation and demethylation, and ...
  53. [53]
    KDM3 epigenetically controls tumorigenic potentials of human ...
    Apr 25, 2017 · Loss of the polycomb mark from bivalent promoters leads to activation of cancer-promoting genes in colorectal tumors. Cancer Res. 74, 3617 ...
  54. [54]
    Identical twins: epigenetics makes the difference - Nature
    Aug 10, 2005 · For 65% of the pairs the twins had almost identical epigenetic profiles, but for the remaining 35% there were significant differences.
  55. [55]
    Epigenetic drift in aging identical twins - PNAS
    Whereas young identical twin pairs are essentially indistinguishable in their epigenetic markings, older identical twin pairs show substantial variations.
  56. [56]
    DNMT1 regulates human erythropoiesis by modulating cell cycle ...
    May 8, 2024 · Here we found that DNMT1 deficiency led to cell cycle arrest of erythroid progenitors which was partially rescued by treatment with a p21 ...
  57. [57]
    Polycomb complexes repress developmental regulators in murine ...
    Here we show that PcG proteins directly repress a large cohort of developmental regulators in murine ES cells, the expression of which would otherwise promote ...
  58. [58]
    Targeting of De Novo DNA Methylation Throughout the Oct-4 Gene ...
    Apr 1, 2010 · Differentiation of embryonic stem (ES) cells is accompanied by silencing of the Oct-4 gene and de novo DNA methylation of its regulatory region.
  59. [59]
    Role of TET enzymes in DNA methylation, development, and cancer
    The ten eleven translocation (TET) enzymes oxidize 5-methylcytosines (5mCs) and promote locus-specific reversal of DNA methylation.
  60. [60]
    The SWI/SNF ATP-dependent chromatin remodeling complex in cell ...
    The SWI/SNF complexes utilize energy from ATP hydrolysis to disrupt histone-DNA interactions and shift, eject, or reposition nucleosomes making the underlying ...
  61. [61]
    The pioneer factor OCT4 requires the chromatin remodeller BRG1 to ...
    We discover that the pluripotency-associated pioneer factor OCT4 binds chromatin to shape accessibility, transcription factor co-binding, and regulatory ...
  62. [62]
    H2A.Z Facilitates Access of Active and Repressive Complexes to ...
    We show that H2A.Z is highly enriched at promoters and enhancers and is required for both efficient self-renewal and differentiation of murine ESCs.
  63. [63]
    PI3Kβ-regulated β-catenin mediates EZH2 removal from promoters ...
    Oct 11, 2022 · β-Catenin deposition at promoters triggers release of the EZH2 repressor, permitting stemness maintenance (through control of NODAL) and correct ...
  64. [64]
    p300 and PCAF Act Cooperatively To Mediate Transcriptional ...
    The transcriptional activity of p300 requires acetyl coenzyme A, indicating that it functions as a histone acetyltransferase when mediating Notch IC function.
  65. [65]
    p300 Acts as a Transcriptional Coactivator for Mammalian Notch-1
    Protein-protein interaction assays demonstrated that the intracellular part of Notch-1 (Notch-1-IC) is targeted by the common coactivator p300.
  66. [66]
    Substratum-induced differentiation of human pluripotent stem cells ...
    We show that matrix mechanics alone can robustly induce neuronal differentiation of hPS cells, independent of soluble neurogenic factors.
  67. [67]
    Matrix stiffness regulation of integrin-mediated mechanotransduction ...
    Osteogenic differentiation was increased on stiffer matrices, as evident by type I collagen, osteocalcin, and Runx2 gene expressions and alizarin red S staining ...
  68. [68]
    Extracellular matrix stiffness controls osteogenic differentiation ... - NIH
    Mar 1, 2018 · Osteogenic differentiation was increased on 62–68 kPa ECM, as evidenced by alpha-1 type I collagen, BGLAP, and RUNX2 gene expression, calcium ...
  69. [69]
    DNA methylation, an epigenetic mechanism connecting folate ... - NIH
    It has been hypothesized that inhibition of methyl transfer or reduced folate intake could increase the risk of human neural tube defects by reducing DNA ...
  70. [70]
    Epigenetic Profiles in Children with a Neural Tube Defect; A Case ...
    Folate deficiency is implicated in the causation of neural tube defects (NTDs). The preventive effect of periconceptional folic acid supplement use is ...
  71. [71]
    HIF‐1α/Actl6a/H3K9ac axis is critical for pluripotency and lineage ...
    Feb 28, 2020 · Meanwhile, hypoxia has appeared as a key feature of the stem cell niche,11, 12 and is beneficial for hESCs, adult stem cells,13-15 and cancer ...
  72. [72]
    Insight into Hypoxia Stemness Control - MDPI
    Hypoxia-inducible factor (HIF) is the master regulator of O2 homeostasis with hundreds of hypoxia-inducible target genes. HIF is a heterodimeric transcription ...
  73. [73]
  74. [74]
    An orderly retreat: Dedifferentiation is a regulated process - PMC - NIH
    Dedifferentiation is the progression of cells from a more differentiated to a less differentiated state. It is observed in a variety of processes such as cancer ...
  75. [75]
    Cell Dedifferentiation - an overview | ScienceDirect Topics
    Cell dedifferentiation is defined as the process in which cells undergo remarkable changes in gene expression, transitioning from a specialized function to ...
  76. [76]
    Lens regeneration: a historical perspective - PMC - PubMed Central
    Adult newts regenerate their lenses through transdifferentiation of cells at the edge of the dorsal iris, but not the ventral part of this tissue. (B) The ...
  77. [77]
    A novel role of the hedgehog pathway in lens regeneration
    Mar 15, 2004 · Lens regeneration in the adult newt begins with proliferation and dedifferentiation of dorsal iris pigment epithelial cells (PECs).
  78. [78]
    Skeletal muscle dedifferentiation during salamander limb regeneration
    A critical step during limb regeneration is formation of a blastema, which gives rise to the new extremity. Salamander limb regeneration has historically been ...
  79. [79]
    Salamander limb regeneration involves the activation of a ...
    Jan 30, 2006 · This process is generally referred to as the dedifferentiation step leading to the formation of blastema progenitors (Iten and Bryant, 1973).
  80. [80]
    DNA Methylation Dynamics Regulate the Formation of a ...
    Elucidation of the specific epigenetic modifications that mediate cellular dedifferentiation likely will lead to insights for initiating a regenerative response ...Missing: reduced | Show results with:reduced
  81. [81]
    How does Lin28 let-7 control development and disease? - PMC
    Jul 9, 2012 · Upon injury several pluripotency factors including c-Myc and Lin28 are rapidly upregulated in response to the dedifferentiation transcription ...
  82. [82]
    Myc-dependent dedifferentiation of Gata6+ epidermal cells ... - Nature
    Sep 21, 2023 · Abstract. Dedifferentiation is the process by which terminally differentiated cells acquire the properties of stem cells. During mouse skin ...Missing: examples | Show results with:examples
  83. [83]
    Cardiac fibroblasts in heart failure and regeneration - Frontiers
    Apr 17, 2024 · Upon cardiac injury, rather than regenerating, mammalian hearts develops chronic fibrosis, a response which is unfortunately maladaptive as it ...
  84. [84]
    Cardiac Fibroblasts and the Extracellular Matrix in Regenerative and ...
    Aug 20, 2019 · In the adult mammalian heart, a cardiac injury results in the formation of a permanent fibrotic scar in the absence of regeneration. In contrast ...
  85. [85]
    Methods for making induced pluripotent stem cells: reprogramming ...
    Feb 22, 2011 · Pluripotent stem-cell lines can be obtained through the reprogramming of somatic cells from different tissues and species by ectopic expression of defined ...
  86. [86]
    Integration-Free Methods for Generating Induced Pluripotent Stem ...
    Induced pluripotent stem (iPS) cells can be generated from mouse or human fibroblasts by exogenous expression of four factors, Oct4, Sox2, Klf4 and c-Myc, and ...
  87. [87]
    Epigenetics of reprogramming to induced pluripotency - PMC
    Evidence is emerging that epigenetic priming events early in the process may be critical for pluripotency induction later.
  88. [88]
    Ezh2 mediated H3K27me3 activity facilitates somatic transition ...
    Feb 4, 2015 · In the current study, we demonstrate that histone methyl transferase activity of Ezh2 is required for mesenchymal to epithelial transition (MET) ...
  89. [89]
    Modeling Parkinson's Disease Using Induced Pluripotent Stem Cells
    In this review, we summarized the advances in molecular mechanisms of Parkinson's disease using iPSC models.
  90. [90]
    Modelling Parkinson's Disease: iPSCs towards Better ... - MDPI
    In this review, we report the available data on iPSC-derived neurons derived from PD patients with identified gene mutations.
  91. [91]
    Autologous Induced Stem-Cell–Derived Retinal Cells for Macular ...
    Mar 16, 2017 · We assessed the feasibility of transplanting a sheet of retinal pigment epithelial (RPE) cells differentiated from induced pluripotent stem cells (iPSCs)
  92. [92]
    Phase I/II trial of iPS-cell-derived dopaminergic cells for Parkinson's ...
    Apr 16, 2025 · Transcriptomics analysis of human iPSC-derived dopaminergic neurons reveals a novel model for sporadic Parkinson's disease ... application of hfVM ...
  93. [93]
    Emergence and evolution of heterocyte glycolipid biosynthesis ...
    Nitrogen-fixing (diazotrophic) cyanobacteria play a major role in nitrogen cycling by transforming N2 to biologically available NH4+ (1–4). To overcome the ...
  94. [94]
    The evolutionary diversification of cyanobacteria: Molecular ... - PNAS
    Apr 4, 2006 · Most cyanobacteria capable of aerobic N2 fixation produce heterocysts (33), whereas a variety of unicellular and simple filamentous forms can ...
  95. [95]
    Cold climate adaptation is a plausible cause for evolution of ... - Nature
    May 29, 2020 · We inferred a fossil-calibrated phylogeny of Dictyostelia, which showed that its two major branches diverged 0.52 billion years ago, following ...
  96. [96]
    The transcription factor Spores Absent A is a PKA dependent ...
    Apr 27, 2018 · Sporulation in Dictyostelium fruiting bodies evolved from amoebozoan encystation with both being induced by cAMP acting on PKA, ...
  97. [97]
    Evolution of individuality during the transition from unicellular to ...
    May 15, 2007 · Koufopanou (24) argued for the volvocine green algae that soma evolved to keep larger colonies afloat and motile while reproductive cells divide ...
  98. [98]
    The evolution of convex trade-offs enables the transition towards ...
    Jul 9, 2021 · The second step in the evolutionary transition toward multicellularity, the soma-germ separation, is more likely expected when the trade-off ...
  99. [99]
    Origin of metazoan cadherin diversity and the antiquity of the ... - PNAS
    Jul 25, 2012 · Our finding of a cadherin gene in C. owczarzaki reveals that cadherins predate the divergence of the C. owczarzaki, choanoflagellate, and metazoan lineages.
  100. [100]
    Hox gene expression determines cell fate of adult periosteal stem ...
    Mar 25, 2019 · Hox genes are evolutionarily conserved transcription factors that during embryonic development function as master regulators of positional ...Missing: conservation species
  101. [101]
    Uncoupled evolution of the Polycomb system and deep origin of non ...
    Nov 10, 2023 · Concurrent conservation of both complexes was most frequent in land plants and animals, while other major taxa displayed a more diverse ...
  102. [102]
  103. [103]
  104. [104]