Fact-checked by Grok 2 weeks ago

Germ cell

Germ cells are the specialized reproductive cells in multicellular organisms that develop into gametes—sperm in males and oocytes in females—thereby enabling and the intergenerational transmission of genetic material. In vertebrates, including mammals, these cells originate as germ cells (PGCs) during early embryogenesis, typically around the time of , where they are specified through inductive signaling from extra-embryonic tissues rather than preformed determinants. PGCs then undergo active migration via the embryonic to the nascent gonadal ridges, a guided by like SDF-1 and c-Kit signaling, before proliferating mitotically and committing to meiotic in response to gonadal cues. Distinct from diploid cells, mature germ cells are haploid following , preserving genomic integrity through unique epigenetic that erases parental imprints to prevent accumulation of deleterious mutations across generations. Disruptions in germ cell specification or migration can lead to infertility or germ cell tumors, underscoring their critical role in reproductive fitness and species continuity.

Introduction and Definition

Biological Role and Characteristics

Germ cells constitute the specialized lineage responsible for gamete production in sexually reproducing , transmitting genetic and epigenetic information across generations to ensure species . As the sole cellular mediators of , they originate from germ cells (PGCs), which are segregated early in embryogenesis from somatic precursors, thereby maintaining a continuous distinct from the mortal body. This segregation preserves the potential for generating totipotent zygotes upon gamete fusion, underpinning organismal renewal and evolutionary adaptation through . In their developmental trajectory, germ cells initially proliferate mitotically as diploid cells before entering meiosis within the gonads to yield haploid gametes—spermatozoa in males and oocytes in females. Meiosis introduces genetic diversity via homologous recombination and independent assortment, halving the chromosome number while facilitating repair of DNA damage accumulated in the parental germline. This process is sex-specific: in females, oogonia arrest in prophase I until puberty or later, whereas male prospermatogonia resume mitosis before meiotic commitment, enabling continuous spermatogenesis. A defining characteristic of germ cells is their retention of totipotent potential, suppressed during somatic restriction but reactivated post-fertilization to direct embryonic . Unlike cells, which terminally differentiate and contribute to organismal function without propagating the , germ cells evade somatic programs through transcriptional repression, reliance on niche signals, and regulatory networks involving factors like Vasa and Nanos. PGCs exhibit morphological hallmarks such as large size, prominent nucleoli, and high activity, alongside molecular markers including OCT4, BLIMP1, and PRDM14, which reinforce their undifferentiated state and migratory competence. These features collectively safeguard integrity against mutational accumulation, prioritizing fidelity in intergenerational transmission over individual somatic longevity.

Distinction from Somatic Cells

Germ cells, also known as germline cells, are the specialized precursors to gametes (sperm and ova) that transmit genetic information across generations, in contrast to cells, which constitute the non-reproductive tissues and organs of the body. This fundamental dichotomy, first articulated by in the late through his germ plasm theory, posits a strict separation where the maintains continuity and heritability while the supports organismal function but does not contribute to inheritance. Early in embryonic development, primordial germ cells (PGCs) are segregated from somatic lineages, often as early as the blastula stage in vertebrates, ensuring that germline cells avoid the somatic differentiation program. A core distinction lies in their proliferative mechanisms and : somatic cells replicate via , preserving diploid number (46 in humans) for tissue maintenance and growth, whereas germ cells transition to in later stages, reducing to haploid (23 chromosomes) to enable and formation. This meiotic process in germ cells introduces variability through crossing over and independent assortment, absent in somatic , which prioritizes fidelity for cellular . Epigenetically, germ cells undergo extensive , including global to erase somatic marks and restore totipotency, allowing them to generate a complete upon fertilization; somatic cells, conversely, accumulate stable epigenetic modifications that lock in differentiated states, rendering them multipotent at best but incapable of full without experimental intervention. The germline's "immortality" underscores its evolutionary primacy: germ cell lineages persist indefinitely across generations, evading the of somatic telomere shortening and , whereas somatic cells are mortal, programmed for finite divisions to prevent unchecked proliferation. Classically encapsulated by the , this separation prohibits heritable changes from somatic mutations or adaptations flowing back to the , though recent studies in model organisms like C. elegans and mice have identified limited soma-to-germline signaling via exosomes or , challenging absolute impermeability without undermining the directional dominance of germline transmission.
AspectGerm CellsSomatic Cells
Primary FunctionGamete production and intergenerational genetic transmissionBody structure, maintenance, and physiological processes
Developmental OriginSegregated early from as primordial germ cellsDerived from remaining embryonic cells post-germline specification
PotencyTotipotent; capable of forming entire via fusionDifferentiated; limited to tissue-specific repair and function
Cell DivisionMitosis in early stages, for Exclusively mitosis
Ploidy (Humans)Diploid initially, haploid post-Diploid throughout
Epigenetic DynamicsExtensive erasure and for Progressive restriction and accumulation of marks for stability
LifespanImmortal across generationsFinite; subject to
These distinctions ensure evolutionary fitness by isolating heritable material from somatic wear, with germ cells bearing the genome's fidelity burden.

Evolutionary and Historical Context

Evolutionary Origins Across Metazoans

In the common ancestor of Metazoa, the germline-soma distinction likely emerged as a specialization enabling dedicated gamete production separate from somatic functions, supported by comparative analyses of developmental modes across phyla. This segregation probably involved inductive specification from multipotent cells rather than preformed determinants, as induction predominates in basal lineages and shows broader phylogenetic distribution than germ plasm inheritance. Molecular markers such as vasa and nanos homologs, which associate with germline functions in diverse animals, trace back to pre-metazoan origins but acquired germline-specific roles early in animal evolution, indicating conserved genetic underpinnings despite mechanistic variation. Basal metazoans like Porifera (sponges) exhibit no early primordial germ cell (PGC) segregation; instead, gametes arise late from multipotent somatic lineages such as choanocytes or archaeocytes via inductive cues, lacking distinct structures. Similarly, , represented by adhaerens, show no dedicated , with reproductive cells differentiating from versatile somatic-like cells under environmental , suggesting a primitive, flexible mode without fixed PGCs. In and , specification remains largely inductive, with germline precursors arising from endodermal or interstitial cells via signaling pathways like , though ctenophores display germ plasm-like granules in oocytes, hinting at early experimentation with preformation that did not persist ancestrally. These patterns imply that the metazoan ancestor relied on late, conditional from totipotent or pluripotent progenitors, minimizing costs in simple body plans. Across , preformation evolved convergently in disparate clades, such as (e.g., nematodes with P granules, annelids with polar plasm) and (e.g., with polar granules), often linked to determinate cleavage and maternal cytoplasmic determinants containing RNAs and proteins like Vasa. In contrast, Deuterostomes predominantly retain , as in amphibians (via germinal granules but requiring signals) and mammals (ZGLP1-mediated epigenetic reprogramming), though represent a derived preformation case with bucky ball-organized . Phylogenetic mapping indicates at least three independent origins of : in spiralians, ecdysozoans, and certain chordates, driven by selection for protected, early isolation amid increasing embryo complexity. This underscores that while the concept is ancient, its implementation diversified to balance fidelity against somatic interference, with as the plesiomorphic state enabling adaptability in early metazoans.

Key Discoveries in Germ Cell Theory

August Weismann formulated the germ plasm theory in 1892, proposing that hereditary continuity is maintained through a distinct germ plasm sequestered in germ cells, separate from somatic cells that cannot transmit acquired traits. This theory emphasized an immutable separation between and , with germ plasm serving as the sole vehicle for across generations, challenging Lamarckian ideas prevalent at the time. Weismann's framework, elaborated in his 1893 monograph Das Keimplasma, laid the groundwork for understanding germ cells as a protected lineage dedicated to reproduction rather than organismal adaptation. Cytological observations in the early provided empirical support for localized germ determinants. In avian embryos, Chester H. Swift identified primordial germ cells (PGCs) originating extra-embryonically and circulating via the vasculature before colonizing the gonads, as detailed in his 1914 study on chick development. In , pole cells forming at the posterior blastoderm pole were recognized as PGC precursors, with transplantation experiments by Illmensee and Mahowald in 1974 demonstrating that posterior polar plasm could induce functional germ cells in ectopic sites, confirming the sufficiency of cytoplasmic for specification. Molecular insights into inductive specification emerged in vertebrates, particularly mammals. In mice, fate-mapping studies by Lawson and Hage in 1994 traced PGC origins to a small epiblast around embryonic day 6.25, establishing their early prior to gonadal migration. Key signaling pathways were elucidated in 1999, when Lawson et al. showed that 4 (BMP4) from extra-embryonic tissues is essential for inducing PGC fate in proximal epiblast cells, marking a shift from descriptive to mechanistic understanding of zygotic induction in mammals. These discoveries highlighted divergent strategies—preformistic via maternal in invertebrates and amphibians versus inductive in mammals—refining Weismann's theory with causal molecular details.

Primordial Germ Cell Specification

Preformation Versus Induction Mechanisms

In primordial germ cell (PGC) specification, two primary mechanisms exist: preformation, which relies on maternally inherited cytoplasmic determinants known as , and , which depends on zygotic signaling from surrounding cells. Preformation enables autonomous determination of germ cell fate during early embryonic cleavages, segregating specialized —composed of RNAs, proteins (such as Vasa and Nanos homologs), and mitochondria—into presumptive PGCs, thereby insulating the germline from somatic influences and repressing zygotic transcription initially. This mode predominates in many and select vertebrates, ensuring robust germline segregation but requiring precise localization of determinants during . Induction, in contrast, specifies PGCs conditionally through extrinsic inductive cues acting on initially pluripotent cells, without reliance on prelocalized . In vertebrates employing this mechanism, such as mice, signals including bone morphogenetic proteins (BMPs, particularly BMP4 and BMP8b) from extraembryonic visceral , combined with WNT3 from posterior , activate key regulators like Blimp1 () and Prdm14 in epiblast cells around embryonic day 6.25 (E6.25). This process represses genes while promoting markers such as Nanos3 and Dazl, rendering PGCs migratory and proliferative. The distinction between preformation and reflects evolutionary divergence, with considered ancestral across bilaterians and preformation arising convergently in lineages like nematodes (e.g., , via P granules), insects (e.g., , via polar granules), and anamniote vertebrates (e.g., with bucky ball-organized ). In preformation systems, of disrupts PGC formation, whereas in -dominant mammals, genetic disruption of or WNT pathways abolishes specification, highlighting causal reliance on somatic-zygotic interactions. modes occur in some species, such as where both and signals contribute, but pure prevails in eutherian mammals, potentially linking to and epigenetic flexibility. These mechanisms ensure germline fidelity but differ in vulnerability: preformation resists environmental perturbation via inheritance, while allows adaptive plasticity at the cost of signaling precision.

Specification in Invertebrates

In many invertebrates, primordial germ cell (PGC) specification occurs via preformation, whereby maternally deposited cytoplasmic determinants, collectively termed germ plasm, are asymmetrically localized in the oocyte and inherited by embryonic cells destined to become the germline.00365-5) This mechanism ensures early segregation of germline fate from somatic lineages through inheritance of RNA-protein complexes that repress somatic differentiation and promote germline-specific gene expression.00496-8) Germ plasm components, including proteins like Vasa and Tudor-domain proteins, form electron-dense granules visible under electron microscopy and are conserved across species employing this strategy. In , germ plasm assembles progressively during oogenesis at the posterior pole of the oocyte, driven by the localization of oskar mRNA, which encodes a protein that nucleates germ plasm formation by recruiting downstream effectors such as nanos, pumilio, and mitochondrial factors. Upon fertilization, this germ plasm induces cellularization of pole cells at the posterior blastoderm margin around 2-3 hours post-fertilization at 25°C, marking the initial PGCs; these cells remain transcriptionally quiescent while repressing somatic genes via Nanos-mediated translational control.00496-8) Experimental ablation of germ plasm, such as through oskar mutants, results in sterility due to failure of pole cell formation, confirming its deterministic role. In the nematode , specification relies on P granules, maternally synthesized ribonucleoprotein aggregates containing proteins like PGL-1 and GLH-1, which localize to the posterior cortex of the via microtubule-dependent transport. Through four rounds of unequal divisions starting at the 2-cell stage, P granules segregate exclusively to the precursor P4 cell by the 16- to 28-cell stage, enforcing germline fate while excluding determinants; mutants lacking P granule components, such as pgl-1, exhibit progressive germline loss across generations. This inheritance maintains germline totipotency amid reprogramming. Although preformation dominates in invertebrates, some species, such as the Helobdella robusta and certain like , employ zygotic inductive signals or hybrid mechanisms, where is absent or insufficient, and PGC fate requires embryonic transcription of zygotic genes responsive to extracellular cues.00365-5) These variations highlight evolutionary flexibility, yet inheritance remains the ancestral and prevalent mode in ecdysozoans.

Specification in Vertebrates

In vertebrates, primordial germ cells (PGCs) are predominantly specified through inductive mechanisms, where extrinsic signaling from neighboring tissues instructs unspecified precursor cells to adopt the germ line fate, contrasting with the preformistic inheritance of seen in many . This process occurs during early or equivalent stages, relying on (BMP) signaling and other pathways to activate germline-specific transcription factors in competent cells of the epiblast or its homologs. In mammals, such as the , PGC specification takes place around embryonic day 6.25 (E6.25) in the proximal epiblast at the junction with extraembryonic tissues. Bone morphogenetic proteins BMP4 and BMP8b, secreted from the extraembryonic visceral , initiate the response by binding to receptors on epiblast cells, leading to the upregulation of core regulators including Blimp1 (encoded by ), Prdm14, and Tfap2c. These factors repress and promote a pluripotency-like state permissive for commitment, with approximately 40-50 PGCs emerging by E7.5. Inhibition of BMP signaling, as shown by studies, abolishes PGC formation, confirming its necessity.00274-8) Amphibians like Xenopus laevis employ a similar inductive strategy, where PGC precursors in the marginal zone receive signals from vegetal cells during blastula stages. BMP signaling, combined with activin/nodal and Wnt pathways, induces expression of germline determinants such as dead end 1 (dnd1) and nanos-like genes by stage 10 (early gastrula). Classical experiments, including UV irradiation of vegetal cytoplasm to disrupt induction, demonstrate that germ plasm-like aggregates are secondary to signaling rather than primary determinants, with rescue possible via transplantation of inducing tissues. In teleosts such as , specification occurs around 4-5 hours post-fertilization (hpf) in the yolk syncytial layer-adjacent blastoderm margin, driven by localized 2b/4 and downstream effectors that activate buckeye (buck), tdrd7, and dnd1. The number of specified PGCs is tightly regulated to about 20-30 per , with ectopic BMP expression expanding the germ cell population, underscoring the instructive role of gradients. Avian species, exemplified by the , specify PGCs inductively in the anterior epiblast shortly after hypoblast formation ( HH3-4), influenced by signals from the and posterior marginal , including and (FGF) pathways that trigger Dazl and Nanos3 expression. Unlike mammalian models, avian PGCs initially disperse widely before concentrating in the germinal crescent, but the core inductive logic persists without evidence of maternally inherited determinants. Across these s, conserved features include the transient pluripotency state of early PGCs and the role of RNA-binding proteins like Dnd1 in protecting germline transcripts, though species-specific variations in timing and spatial cues reflect adaptations to diverse developmental architectures. Experimental manipulations, such as overexpression or receptor mutants, consistently validate as the dominant mode, with no vertebrate models relying solely on preformation in the surveyed systems.

Migration to Gonadal Ridges

Migration Mechanisms in Invertebrates

In , germ cells (PGCs) originate at the posterior pole of the early embryo (embryonic stages 4–5, approximately 1.5–3 hours after egg laying) and are passively incorporated into a pocket during (stage 9, ~4 hours after egg laying). Active migration commences with transepithelial from the epithelium (stages 9–10, ~4.5 hours after egg laying), followed by reorientation toward the dorsal (stage 10, ~5 hours after egg laying) and bilateral dispersal to gonadal precursors (SGPs) in parasegments 10–12 (stage 11, ~7 hours after egg laying), culminating in coalescence. Migration relies on amoeboid driven by at the , rear actomyosin contractility, and continuous cortical flows that maintain rounded shape and adaptability to substrates. Guidance integrates repulsive and attractive cues: lipid phosphate phosphatases Wunen and Wunen-2 in the degrade phospholipids to generate repulsive gradients, deterring ventral retention and promoting survival by limiting competition for attractants; conversely, SGPs produce lipid chemoattractants via the (HMGCR) pathway, including geranylgeranylated proteins and isoprenoids. The Tre1 in PGCs detects these cues, coupling to heterotrimeric G proteins to activate Rho1 , which polarizes the and redistributes E-cadherin for epithelial exit and substrate interactions. and are further supported by JAK/STAT and /MAPK signaling from somatic tissues, while the Mdr49 exports attractants from SGPs. Juvenile hormones, synthesized via the , act locally in embryos to direct PGCs to SGPs, with mutants showing defective coalescence as of studies published in 2024.01733-5) In , PGCs (Z2 and Z3) undergo minimal active migration, instead internalizing during through E-cadherin-dependent adhesions that enable "hitchhiking" on endodermal progenitors, followed by displacement via somatic rearrangements rather than long-range . This contrasts with the cue-directed, invasive motility in , highlighting species-specific adaptations in nematode gonad primordium formation.

Migration Pathways and Cues in Vertebrates

In vertebrates, primordial germ cells (PGCs) migrate from early embryonic specification sites to gonadal ridges through diverse pathways shaped by embryonic anatomy, involving passive translocation during followed by active, directed motility via and substrate interactions. The stromal cell-derived factor 1 (SDF-1, also known as ) and its receptor form a conserved , creating gradients that attract CXCR4-expressing PGCs toward targets expressing SDF-1, with disruptions causing mislocalization or failure to colonize gonads. Additional cues, such as for and KIT ligand (KITL) for survival and motility, support this process, while PGCs employ amoeboid with bleb protrusions driven by dynamics and Rho . In teleost fish such as , PGCs originate at the vegetal margin of the blastoderm and undergo a multi-phase : initial dorsal convergence during shield stage, followed by anterior-lateral traversal through and to the dorsal and gonadal primordia by 24 hours post-fertilization (hpf). This active path relies on dynamic SDF-1a gradients regulated by somatic CXCR7b-mediated internalization, prompting "run-and-tumble" bleb-based where leading-edge blebs form via localized and contractility. Supporting factors include for prenylation-dependent G-protein signaling and signaling for survival, with maternal dead end mRNA suppressing to maintain migratory competence. Amphibian PGCs, as in Xenopus laevis, are passively incorporated into the during before actively migrating dorsally through and the dorsal mesentery to gonadal ridges, utilizing for substrate probing on matrices. SDF-1/ signaling directs this later phase, with knockdowns impairing dorsal navigation and survival; reduced E-cadherin promotes from endodermal clusters, enabling elongated morphology with RhoA-mediated rear contraction and PIP3-enriched leading blebs. Approximately 30 PGCs typically arrive per , with integrity preventing ectopic invasion. In birds like chickens, PGCs migrate via a vascular route: originating in the germinal crescent, they enter hypoblastic blood islands by stage X (early ), circulate through the bloodstream and , then at the hindgut region before traversing the dorsal to gonads. SDF-1 guides vascular exit and mesentery navigation, complemented by and biasing rightward asymmetry; store-operated calcium entry (SOCE) drives bleb formation during extravasation. Mammalian PGCs, exemplified by mice, emerge in the proximal epiblast around embryonic day 6.5 (E6.5), ingress through the posterior into by E7.5, then actively migrate laterally through the to s by E10.5-E11.5, expanding from ~40 to thousands of cells. SDF-1 from ridges is essential for homing, with mutants failing colonization despite earlier motility; KITL-Kit signaling enhances and Rac1-dependent speed, while β1-integrins and provide traction, and passively aligns PGCs. Asynchronous arrival ensures colonization even if some PGCs stray. Across vertebrates, early routes diverge—tissue-based in and amphibians, bloodstream-dominant in and reptiles—but converge on the dorsal mesentery for final chemotactic homing, underscoring evolutionary conservation of SDF-1/ amid adaptive variations. Defects in these cues lead to sterility via , as PGCs undergo without successful integration.

Epigenetic Reprogramming in Germ Cells

Erasure of Somatic Epigenetic Marks

In primordial germ cells (PGCs), the erasure of epigenetic marks constitutes a critical phase of epigenetic reprogramming, involving the systematic removal of patterns and repressive modifications inherited or acquired from lineages. This process resets the epigenome, eliminating cellular memory and enabling the acquisition of germline-specific states essential for totipotency and . In mammals, this erasure begins shortly after PGC specification and intensifies during migration to the gonadal ridges, ensuring that epimutations or aberrant imprints are not propagated to offspring.00932-X) The primary epigenetic mark targeted is at CpG sites, which undergoes genome-wide demethylation. In mice, PGCs exhibit initial methylation levels comparable to cells (approximately 70% CpG methylation at E6.5), followed by loss: partial reduction by E9.5, and near-complete (<10% ) by E13.5 across genic, intergenic, and repetitive regions, including most CpG islands. This hypomethylation affects thousands of loci associated with developmental regulators, transposons, and metabolic genes, though certain multicopy sequences or evolutionarily young retroelements may retain partial . In humans, analogous occurs between weeks 7-11 post-fertilization, with similar depletion but distinct kinetics influenced by extended PGC proliferation.00932-X) Mechanisms of DNA demethylation combine active and passive pathways. Active demethylation is mediated by TET1 and TET2 enzymes, which oxidize 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and further intermediates, facilitating base excision repair or replication-independent removal; TET1 expression peaks in migrating mouse PGCs around E8.5-E11.5. Passive demethylation predominates due to proliferation without efficient maintenance methylation, driven by transient downregulation of UHRF1 and DNMT1 from E7.5 onward, coupled with upregulated replication. These processes are interdependent, with chromatin remodeling (e.g., H3K27me3 deposition) preceding and facilitating demethylation at select loci. Experimental disruption, such as TET1 knockout, impairs demethylation at germline-specifying genes but not globally, indicating redundancy. Beyond DNA methylation, somatic histone modifications are erased, notably H3K9me2 from pericentromeric heterochromatin and broad H3K27me3 domains established in somatic progenitors. In mouse PGCs, H3K9me2 loss initiates at E8.0, correlating with transcriptional activation and nuclear reorganization, while genome-wide profiling reveals depletion of somatic-enriched marks like H3K27ac at lineage genes. These changes, observed via ChIP-seq, restore a more open chromatin landscape akin to embryonic stages, though not all marks (e.g., some H3K4me3) persist to guide demethylation. In vitro PGC-like cell models confirm this erasure recapitulates in vivo dynamics, with incomplete removal linked to epimutation retention. This erasure is not uniform; somatic imprints at imprinted genes are erased later (E10.5-E13.5 in mice), but broad somatic patterns are preferentially targeted early to avert intergenerational defects. Failures in erasure, as in mutants, lead to retained somatic methylation, infertility, or teratoma formation, underscoring its role in safeguarding germline integrity. Across metazoans, conserved elements like TET homologs suggest evolutionary robustness, though avian PGCs show reinforced repression before demethylation.

Re-establishment of Germline-Specific Imprints

Following the erasure of somatic epigenetic marks in primordial germ cells (PGCs), germline-specific genomic imprints—primarily differential DNA methylation at imprinting control regions (ICRs)—are re-established de novo during gametogenesis in a sex-specific manner to ensure parent-of-origin-dependent gene expression in the offspring. This re-establishment restores monoallelic expression for approximately 100-200 imprinted genes in mammals, with maternal imprints acquired in oogenesis and paternal imprints in spermatogenesis; failure in this process can lead to imprinting disorders such as or . The process relies on DNA methyltransferases (DNMTs), particularly and its cofactor , which target germline differentially methylated regions (gDMRs) for allele-specific methylation. In the female germline, maternal imprints are established progressively during oocyte growth within ovarian follicles, beginning after birth in mice (around postnatal day 0-3) and extending through the growth phase until completion by the fully grown oocyte (FGO) germinal vesicle stage. Of the known gDMRs, 16 acquire methylation in oocytes, marking paternal alleles for silencing post-fertilization; this occurs asynchronously across loci, with some like Igf2r methylated early in growth and others like Peg3 later, influenced by local chromatin features such as H3K36me3 and transcription through the region. DNMT3L recruits DNMT3A to these sites, and genetic ablation of either enzyme results in imprinting defects and infertility, as demonstrated in knockout mouse models where oocyte-derived embryos exhibit biallelic expression of maternal imprints. In humans, similar timing aligns with fetal and postnatal oocyte development, though direct studies are limited by ethical constraints. In the male germline, paternal imprints form earlier and more uniformly, initiating in prospermatogonia at embryonic day 14.5 in mice, prior to meiosis, and completing in perinatal prospermatogonia with maintenance through subsequent mitotic divisions. Only three gDMRs (H19/Igf2, Meg3, Rasgrf1) gain methylation in spermatogenesis, targeting maternal alleles; this process also depends on DNMT3A/DNMT3L but occurs in a post-migratory, mitotic context without the prolonged growth phase seen in oogenesis. Transcription across ICRs is required for accessibility, as evidenced by studies showing delayed or absent methylation in mutants lacking RNA polymerase II activity at these loci. Unlike oogenesis, male imprint establishment precedes gonadal sex differentiation completion, ensuring robustness against earlier epigenetic volatility in PGCs. These sex-dimorphic patterns reflect evolutionary adaptations for gamete production timelines—protracted in females for resource accumulation versus rapid in males—and are conserved across eutherian mammals, with evidence from comparative epigenomic profiling in mice, humans, and cattle confirming gDMR orthology and methylation dynamics. Disruptions, such as environmental exposures or mutations in DNMT3L, underscore the precision required, as partial imprint failure correlates with embryonic lethality or growth phenotypes in model organisms.

Gametogenesis Processes

Overview of Meiosis and Differentiation

Meiosis represents the reductive division essential for germ cell development, converting diploid primordial germ cells (PGCs) or their mitotic descendants into haploid gametes while facilitating genetic recombination. Following mitotic proliferation and epigenetic reprogramming, germ cells undergo a pre-meiotic S phase, replicating DNA to yield a 4C DNA content with paired sister chromatids per chromosome. This sets the stage for two successive divisions: meiosis I, which segregates homologous chromosomes, and meiosis II, which separates sister chromatids, ultimately producing four haploid cells from one diploid precursor. In meiosis I, prophase I dominates, encompassing leptotene (chromosome condensation), zygotene (synapsis via synaptonemal complex formation), pachytene (crossing over between non-sister chromatids, mediated by Spo11-induced double-strand breaks), diplotene (partial desynapsis), and diakinesis (further condensation). Metaphase I aligns bivalents at the equator, followed by anaphase I's homolog segregation and telophase I's cytokinesis, yielding two secondary germ cells with haploid chromosome sets but duplicated chromatids. Meiosis II mirrors mitosis: prophase II lacks recombination, while metaphase II, anaphase II, and telophase II distribute chromatids into four haploid nuclei, with cytokinesis completing gamete formation. Recombination during prophase I, averaging 1-3 crossovers per chromosome pair in humans, ensures allelic shuffling and proper segregation via chiasmata. Differentiation integrates with meiosis, transforming post-meiotic haploid cells into functional gametes through sex-specific morphological and biochemical changes. In this phase, transcriptional reprogramming activates genes for cellular remodeling, such as acrosome formation or cytoplasmic expansion, while suppressing somatic traits to preserve germline integrity. Timing of meiotic entry—embryonic in female gonads via retinoic acid signaling, post-pubertal in males—commits germ cells to gamete fates, with failures risking aneuploidy or sterility. These processes halve ploidy to restore diploidy upon fertilization, maintaining species genome stability across generations.

Sex-Specific Adaptations

In mammalian germ cells, entry into meiosis exhibits pronounced sex-specific timing regulated by gonadal somatic signals. Female primordial germ cells (PGCs) transition to oogonia and initiate meiosis during fetal development, progressing to prophase I before arresting in the dictyate stage, which persists from birth until ovulation—potentially spanning decades in humans. Male PGCs, in contrast, proliferate mitotically as prospermatogonia during fetal and early postnatal life, delaying meiotic entry until puberty under retinoic acid signaling from Sertoli cells, enabling continuous spermatogenesis throughout adulthood. This temporal divergence ensures female germ cells align with limited ovarian follicle reserves, while male adaptations support high-volume sperm production exceeding 100 million daily in humans. Meiotic progression further diverges in checkpoint mechanisms and chromosomal handling. Females exhibit a less stringent spindle assembly checkpoint (SAC), permitting higher aneuploidy rates in oocytes—observed in up to 20-25% of human eggs—which may reflect adaptations for maternal age-related selection via embryonic lethality rather than stringent pre-meiotic filtering. Males enforce tighter SAC controls, particularly to resolve X-Y chromosome pairing via pseudoautosomal regions during pachytene, minimizing sex chromosome aneuploidy in sperm, which rarely exceeds 1-2%. Genome organization also differs: mouse spermatocytes cluster telomeres peripherally for efficient recombination, whereas oocytes maintain more dispersed configurations, potentially adapting to prolonged prophase for DNA repair. Cytokinesis and gamete output represent asymmetric adaptations in females versus symmetric in males. Oogenesis yields one large oocyte and three polar bodies through unequal divisions, concentrating cytoplasmic resources for embryonic support, with meiosis II completing only post-fertilization. Spermatogenesis produces four equivalent haploid spermatids, optimized for motility and quantity over provisioning, without post-meiotic arrest. These features correlate with differential stress responses; female germ cells resist glucocorticoid-induced apoptosis via intrinsic buffering, preserving limited oocyte pools, while male cells remain susceptible to enhance turnover. Such adaptations underpin reproductive dimorphism, with female mechanisms prioritizing quality amid scarcity and male processes favoring proliferation despite elevated mutation risks from extensive divisions.

Oogenesis

Stages from Primordial Germ Cells to Oocytes

In the developing female gonad, primordial germ cells (PGCs) that have migrated from the yolk sac differentiate into oogonia upon receiving signals from somatic cells in the ovarian stroma, marking the onset of oogenesis. This transition occurs in humans around gestational weeks 5 to 6, with oogonia characterized by their diploid (2N) DNA content and continued mitotic potential. Oogonia initially reside in germ cell nests, clusters formed by incomplete cytokinesis during proliferation, which provide a protective microenvironment influenced by factors such as KIT ligand from surrounding mesenchyme. Oogonia then enter a phase of rapid mitotic proliferation, expanding the germ cell pool to support the high oocyte demand in mammals. In humans, this multiplication peaks between gestational weeks 8 and 20, generating up to 6-7 million oogonia before significant attrition begins. Proliferation is regulated by paracrine signals including retinoic acid and bone morphogenetic proteins (BMPs), which promote DNA replication while suppressing apoptosis in viable cells. By mid-gestation, approximately half of the oogonial population undergoes programmed cell death, reducing numbers through nest breakdown mediated by somatic cell invasion and germ cell autophagy. The shift from mitosis to meiosis defines the formation of primary oocytes, as oogonia arrest cell division, undergo premeiotic DNA replication to achieve a tetraploid (4N) state, and progress into prophase I of meiosis I. In humans, this entry begins asynchronously around weeks 7-13 of gestation, driven by ovarian-specific retinoic acid gradients that activate STRA8 and other meiotic initiators, distinguishing female germ cells from mitotic spermatogonia in males. Primary oocytes advance to the diplotene stage of prophase I, where homologous chromosomes partially synapse and recombine, establishing chiasmata essential for proper segregation, before arresting in a dictyate state. Arrested primary oocytes become enclosed by flattened pre-granulosa cells derived from the ovarian surface epithelium and mesonephros, forming primordial follicles by gestational weeks 18-20 in humans. This assembly involves selective survival of oocytes post-nest breakdown, with surviving units entering meiotic arrest that persists from fetal life through adulthood, ensuring a reserve for cyclic recruitment at puberty. At birth, the human ovarian pool contains about 1-2 million primordial follicles, reflecting a 65-80% reduction from peak fetal numbers due to atresia.

Oocyte Growth, Meiotic Arrest, and Maturation

Oocyte growth occurs primarily within preantral and antral follicles in the mammalian ovary, where the oocyte accumulates cytoplasmic components essential for embryonic development, including mitochondria, ribosomes, and maternal mRNAs. In humans, the oocyte diameter expands from approximately 20-30 μm in primordial follicles to about 120 μm in fully grown oocytes, accompanied by a four-fold increase in polyribosomes to support protein synthesis demands. This growth phase, lasting months to years depending on species, involves bidirectional communication with surrounding granulosa and theca cells, which provide nutrients and regulatory signals via gap junctions. Meiotic arrest in primary oocytes is established at the diplotene stage of prophase I shortly after birth in mammals, preventing progression until the preovulatory luteinizing hormone (LH) surge. This arrest is maintained by elevated intra-oocyte cyclic adenosine monophosphate (cAMP) levels, generated through constitutive activation of the G-protein-coupled receptor , which stimulates adenylyl cyclase via Gs proteins. Synergistically, cyclic guanosine monophosphate (cGMP) diffuses from cumulus and mural granulosa cells into the oocyte, inhibiting phosphodiesterase 3A (PDE3A) and thereby preserving cAMP; granulosa-derived purines like hypoxanthine and xanthine further suppress PDE3A activity. These mechanisms ensure transcriptional quiescence and cytoplasmic maturation while homologous chromosomes remain paired, minimizing genetic instability over extended arrest periods, which can span decades in humans. Oocyte maturation resumes upon the mid-cycle LH surge, which indirectly signals the oocyte by disrupting granulosa cell cGMP production and gap junctional transfer, leading to PDE3A activation, cAMP hydrolysis, and meiotic resumption. Germinal vesicle breakdown (GVBD) follows within 15 hours in humans, marking progression from prophase I; the oocyte then completes meiosis I, extruding the first polar body, and arrests again at metaphase II of meiosis II, where it awaits fertilization. This process, coordinated with cumulus expansion and follicular rupture, typically culminates 35 hours post-LH surge with polar body emission, ensuring the oocyte achieves fertilizable competence with reduced aneuploidy risk through checkpoint mechanisms.

DNA Repair Mechanisms and Mutation Rates in Oogenesis

Oocytes in mammals, including humans, maintain genomic stability during oogenesis through a suite of DNA repair pathways adapted to the unique challenges of meiotic arrest and prolonged quiescence. Double-strand breaks (DSBs), which arise endogenously from meiotic recombination or spontaneously from oxidative stress and replication errors, are primarily repaired via homologous recombination (HR) in prophase I-arrested oocytes, utilizing the sister chromatid as a homologous template to minimize errors. Experimental induction of DSBs in mouse oocytes reveals efficient HR-mediated repair, achieving near-complete resolution when apoptosis is suppressed, underscoring the pathway's fidelity in preserving oocyte viability. Non-homologous end joining (NHEJ), an alternative DSB repair mechanism, predominates in metaphase II (MII) oocytes, where HR substrates are limited, though its error-prone nature can introduce small insertions or deletions if invoked prematurely. Base excision repair (BER) handles oxidative base lesions, with constitutive activity detected in MII oocytes to counter reactive oxygen species accumulation during follicular growth. Repair efficiency varies by oocyte stage and maternal age. During diplotene arrest, which spans from fetal development to ovulation and can exceed 40 years in humans, local DNA synthesis facilitates precise repair of clustered damage via error-free pathways like HR, supported by dynamic chromatin remodeling and repair foci mobility. However, aged oocytes exhibit diminished repair capacity, characterized by immobilized damage sites, cohesin depletion disrupting repair compartments, and a shift toward NHEJ, correlating with elevated aneuploidy and fragmentation rates observed in women over 35. Autophagy intersects with repair by facilitating RAD51 recruitment to DSBs, mitigating age-related damage escalation, while primordial follicle oocytes retain robust DSB repair for diverse insults, though this wanes post-puberty. Mutation rates in the female germline remain low, reflecting oogenesis's limited proliferative phases—primarily mitotic divisions in fetal oogonia followed by meiotic arrest—coupled with vigilant repair. Human de novo mutation rates average 1.2 × 10^{-8} per base pair per generation, with maternal contributions comprising 20-25%, far below paternal rates due to fewer replication cycles and HR dominance during susceptible periods. Duplex sequencing of single oocytes from women aged 20-42 detects base substitution rates around 10^{-8} to 10^{-7} per nucleotide, without significant maternal age escalation, attributable to allele frequency selection and stringent quality control rather than replication fidelity alone. In contrast to spermatogenesis's continuous divisions, oogenesis yields 3-4-fold fewer mutations across amniotes, as unrepaired lesions trigger atresia rather than propagation. Replication errors during pre-meiotic mitoses contribute detectable de novo variants at rates of 10^{-6} per nucleotide in isolated oocytes, yet overall germline transmission remains constrained by apoptotic culling of defective cells. This asymmetry underscores causal roles of division count and repair pathway bias in shaping sex-specific mutation spectra.

Spermatogenesis

Stages from Spermatogonia to Spermatozoa

Spermatogenesis proceeds through three principal phases—spermatocytogenesis, meiosis, and spermiogenesis—within the seminiferous tubules of the testes, where germ cells interact closely with for structural support and nourishment. This process commences at puberty and continues throughout adult life, producing millions of spermatozoa daily in humans. In spermatocytogenesis, diploid spermatogonia at the tubule base undergo mitotic proliferation to sustain the stem cell reservoir and generate differentiating cells. Type A spermatogonia, including subtypes Ad (dark, reserve stem cells) and Ap (pale, actively renewing), divide asymmetrically to self-renew while producing type B spermatogonia; these type B cells then commit to differentiation, replicate DNA, and transition to preleptotene primary spermatocytes. This phase amplifies germ cell numbers prior to reduction division. The meiotic phase follows, with primary spermatocytes enlarging and entering prophase I, where homologous chromosomes pair, undergo synapsis, and exchange genetic material via crossing over, enhancing diversity. Meiosis I completes to yield haploid secondary spermatocytes (each with 23 duplicated chromosomes), which almost immediately enter meiosis II, dividing equitably to form four round spermatids per original primary spermatocyte. Cytokinesis in later stages is incomplete, linking cells in syncytia supported by Sertoli processes. Spermiogenesis remodels round spermatids into streamlined spermatozoa without further cell division or DNA replication. It encompasses four sequential subphases: the Golgi phase, where proacrosomic vesicles arise from the Golgi complex and coalesce into the nascent acrosome; the cap phase, with the acrosome flattening and enveloping half the nuclear surface; the acrosome phase, featuring nuclear chromatin condensation via histone-protamine replacement, mitochondrial migration to the midpiece, flagellar axoneme elongation, and excess cytoplasm accumulation into a residual body; and the maturation phase, culminating in spermiation as Sertoli cells phagocytose the residual body and release elongated spermatozoa into the tubule lumen. These transformations confer motility, capacitation readiness, and acrosomal enzymes for egg penetration. The complete human spermatogenic cycle spans approximately 64 to 74 days, with one epithelial cycle lasting about 16 days and encompassing 4.5 to 4.6 cycles for full maturation; post-release, spermatozoa undergo further epididymal transit for 10–14 days to gain fertilizing competence.

Spermiogenesis and Sperm Maturation

Spermiogenesis represents the post-meiotic differentiation of haploid round spermatids into mature spermatozoa, occurring within the seminiferous tubules of the testis. This process involves profound morphological remodeling, including acrosome formation, nuclear condensation, flagellar assembly, and cytoplasmic reduction, without further cell division. In mammals, spermiogenesis is classified into four phases based on acrosomal development: Golgi, cap, acrosome, and maturation. During the Golgi phase, the Golgi apparatus in the round spermatid produces proacrosomal granules that coalesce into the acrosomic vesicle, which begins to flatten against the nuclear envelope. Concurrently, the centrosome duplicates, with the distal centriole migrating to form the basis of the flagellum's axoneme, characterized by a 9+2 microtubule arrangement essential for motility. The cap phase follows, where the acrosomic vesicle spreads over the anterior nucleus like a cap, covering 40-50% of its surface, while the spermatid remains spherical. In the acrosome phase, the spermatid elongates as the acrosome further expands and flattens, polarizing the cell with the acrosome and nucleus at one pole and the developing flagellum at the opposite. Nuclear condensation intensifies through histone hyperacetylation, eviction of histones, and replacement with protamines, achieving over 95% chromatin compaction by the end of spermiogenesis to protect paternal DNA. Excess cytoplasm is phagocytosed by Sertoli cells, and the manchette—a transient microtubule structure—guides head shaping and tail elongation. The maturation phase completes flagellar development, with the axoneme extending into a principal piece surrounded by fibrous sheath and mitochondria-rich midpiece for ATP supply. Spermiation then releases elongated spermatids from Sertoli cells into the tubule lumen. Testicular spermatozoa are immotile and infertile at this stage. Post-testicular sperm maturation occurs during 10-14 day transit through the epididymis, where spermatozoa acquire progressive motility and fertilizing competence via region-specific secretory and absorptive activities of principal cells. In the caput epididymidis, surface glycoproteins are modified through glycosylation and shedding, altering membrane properties; the cytoplasmic droplet migrates from the annulus to the midpiece. Proteomic remodeling includes protein secretion and adsorption, enhancing zona pellucida binding affinity. Luminal fluid acidification by proton pumps inhibits premature capacitation, while lipid adjustments prepare for cholesterol efflux in the female tract. Cauda epididymidal spermatozoa exhibit forward motility due to mitochondrial reorganization and axonemal maturation but require final activation via bicarbonate and calcium influx for hyperactivated motility during fertilization. Disruptions in epididymal maturation, such as pH dysregulation, lead to asthenozoospermia.

DNA Repair Mechanisms and Mutation Rates in Spermatogenesis

During spermatogenesis, DNA repair mechanisms are critical to counteract damage from replication errors, oxidative stress, and meiotic recombination, given the continuous mitotic divisions of spermatogonial stem cells and the vulnerability of haploid germ cells. Key pathways active in pre-meiotic stages include base excision repair (BER) for oxidative lesions, nucleotide excision repair (NER) for bulky adducts, and mismatch repair (MMR) to correct replication mismatches, all of which maintain genome integrity across hundreds of cell divisions per lifetime. Double-strand breaks (DSBs), induced deliberately during meiotic recombination in spermatocytes, are primarily repaired via homologous recombination (HR) using sister chromatids or homologs as templates, ensuring proper segregation and crossover formation. In post-meiotic spermatids, repair capacity diminishes as histones are replaced by protamines during spermiogenesis, limiting access to DSB repair factors and potentially leaving unresolved breaks that persist into mature spermatozoa. Alternative DSB repair modes, such as classical non-homologous end joining (cNHEJ), alternative end-joining (aEJ), and single-strand annealing (SSA), may operate in later stages but introduce higher error rates, including insertions, deletions, or loss of heterozygosity, compared to HR. Deficiencies in specific factors, like MCM9 involved in HR, lead to unrepaired DSBs, germ cell depletion, and phenotypes such as Sertoli cell-only syndrome in humans and mice. Oxidative damage from reactive oxygen species (ROS), prevalent due to high metabolic activity in testes, is addressed by BER and antioxidant defenses, but incomplete repair contributes to strand breaks in spermatozoa, which lack robust cytoplasmic repair machinery. Mutation rates in human spermatogenesis reflect the cumulative impact of these processes, with the male germline mutation rate estimated at 1.0–2.0 × 10^{-8} per nucleotide per generation, driven by replication errors accumulating over approximately 23 years of divisions from zygote to conception (versus ~30 total divisions in females). Approximately 80% of de novo mutations in offspring arise paternally, with an average of 50–70 novel single-nucleotide variants per diploid genome, increasing linearly by about 1–2 mutations per additional year of paternal age due to extra mitotic cycles in spermatogonia. This age effect is exacerbated by selfish selection, where clones of spermatogonia harboring advantageous mutations (e.g., in growth-promoting genes) expand preferentially, elevating transmission of certain driver mutations and contributing to disorders like achondroplasia or developmental anomalies. Empirical sequencing of sperm DNA confirms positive selection amplifies specific variants during clonal expansion, though overall fidelity relies on error-prone tolerance in late-stage repair to avoid apoptosis and preserve fertility. Despite these safeguards, unrepaired damage correlates with reduced sperm motility and increased infertility risk, underscoring the trade-off between mutation accumulation and reproductive success.

Pathologies and Diseases

Germ Cell Tumors: Types and Etiology

Germ cell tumors (GCTs) are neoplasms originating from primordial germ cells, which retain pluripotency and can differentiate into various tissue types. They are histologically classified into two broad categories: seminomatous tumors, which resemble undifferentiated germ cells, and non-seminomatous tumors, which exhibit more differentiated or extra-embryonic features. This classification applies across gonadal and extragonadal sites, though prognosis and molecular profiles vary by location. Testicular GCTs predominate in males aged 15–44 years, with an incidence of 6.0 per 100,000 men annually in the United States. Ovarian GCTs are rarer, comprising 2–5% of ovarian malignancies, primarily affecting adolescents and young women. Extragonadal GCTs, arising in midline sites like the mediastinum, retroperitoneum, or central nervous system, account for 2–5% of all GCTs and often present with poorer outcomes due to delayed diagnosis. Seminomatous GCTs include classic seminoma in the testis and its ovarian counterpart, dysgerminoma, both characterized by uniform cells resembling primordial germ cells with lymphocytic infiltrates and syncytiotrophoblasts in some cases. Non-seminomatous GCTs encompass embryonal carcinoma (undifferentiated embryonic-like cells), yolk sac tumor (endodermal sinus structures producing alpha-fetoprotein), choriocarcinoma (trophoblastic elements producing beta-human chorionic gonadotropin), teratoma (mature or immature tissues from three germ layers), and mixed forms combining these elements. Spermatocytic tumors, a rare non-seminomatous variant not derived from intratubular germ cell neoplasia in situ (IGCN), occur almost exclusively in the testis of older men and lack isochromosome 12p. In ovarian cases, immature teratomas are notable for their grading based on neuroectodermal content, while mixed GCTs often include dysgerminoma components. Extragonadal tumors mirror gonadal histology but show higher rates of non-seminomatous subtypes in mediastinal sites. The etiology of GCTs involves disrupted migration or maturation of primordial germ cells during embryogenesis, leading to persistent pluripotency and susceptibility to oncogenic transformation. In testicular GCTs, pathogenesis typically begins with IGCN formation in utero, remaining dormant until puberty when hormonal surges (e.g., gonadotropins) drive progression; nearly all type II TGCTs (seminomas and non-seminomas) arise from this precursor, marked by 12p gain (often i(12p)) and low somatic mutation burden (0.5 per Mb). Genetic predisposition is substantial, with 78 susceptibility loci identified via genome-wide association studies explaining ~44% of heritability; these loci cluster in pathways for germ cell development, sex determination, and chromosomal segregation, with polygenic risk scores conferring up to 6.8-fold increased risk in high-risk quartiles. Key risk factors include cryptorchidism (4- to 8-fold elevated risk, mediated by aberrant germ cell maturation), family history (4- to 10-fold in first-degree relatives), testicular dysgenesis syndrome components like infertility, and genomic variants in genes such as KITLG (odds ratio >2.6). Environmental contributors, including potential endocrine disruptors or use, remain correlative rather than causal, with rising incidence (e.g., among men) suggesting gene-environment interactions. Ovarian GCT etiology is less elucidated but similarly implicates genetic mutations in germ cell precursors, potentially linked to chromosomal abnormalities or inherited syndromes, though specific loci are fewer than in testicular cases. Risk factors overlap with but lack strong familial clustering; environmental factors are hypothesized but unproven. Extragonadal GCTs may stem from misplaced germ cells along migratory routes, with mediastinal non-seminomas showing distinct genomic instability beyond 12p alterations. Overall, GCTs exhibit high (37–49%) yet low , underscoring polygenic and developmental origins over single mutations.

Infertility Syndromes and Genetic Aberrations

Klinefelter syndrome, characterized by a 47,XXY karyotype, results in progressive germ cell depletion beginning at puberty, leading to azoospermia in over 90% of affected adult males and universal infertility without intervention. This condition arises from meiotic nondisjunction, causing disrupted spermatogenesis due to abnormal X-chromosome pairing and Sertoli cell dysfunction. Y-chromosome microdeletions in azoospermia factor (AZF) regions, particularly complete AZFc deletions, represent the most frequent genetic cause of non-obstructive azoospermia or severe oligospermia, impairing spermatogonial proliferation and differentiation. These deletions occur in 10-15% of men with azoospermia and are transmitted via the paternal lineage, though affected males are infertile. Sertoli cell-only syndrome, also termed germ cell aplasia or del Castillo syndrome, manifests as complete absence of germ cells in seminiferous tubules, resulting in and primary testicular failure in approximately 10-25% of infertile males evaluated via . Genetic underpinnings include mutations in genes such as TEX11, which disrupt meiotic recombination and , leading to arrest at stages. Pathogenic variants in germ cell nuclear acidic peptidase (GCNA), essential for during , have been identified in cases of meiotic arrest and non-obstructive . In females, (45,X ) causes ovarian dysgenesis through accelerated primordial germ cell from early fetal stages, yielding streak gonads devoid of functional oocytes and with near-universal . This aberration stems from meiotic errors or postzygotic loss of the second sex chromosome, with residual oocytes rarely surviving beyond infancy. Mutations in meiosis-specific genes, such as those involved in chromosome segregation (e.g., SYCP3 or MSH4), precipitate premature ovarian insufficiency by halting oocyte maturation at I, reducing and . Broader genetic aberrations, including balanced translocations or inversions disrupting meiotic pairing, elevate risks in gametes, contributing to recurrent pregnancy loss or via embryonic arrest. In both sexes, mutations in pathways active in germ cells, such as those rectified during or , amplify mutation rates and meiotic errors, underscoring the causal role of unrepaired double-strand breaks in syndromic . Screening for these aberrations, including karyotyping and targeted sequencing, informs and options like preimplantation in assisted reproduction.

Environmental and Lifestyle Risk Factors

Exposure to endocrine-disrupting chemicals (EDCs), such as (BPA) and , has been linked to disruptions in germ cell (PGC) and , potentially increasing the risk of reproductive disorders including germ cell tumors (GCTs). These compounds, found in plastics and , interfere with hormonal signaling, leading to epigenetic changes in fetal germ cells that may persist transgenerationally, as evidenced by reduced in models exposed to low doses of BPA (0.5–50 µg/kg/day). Human epidemiological data associate prenatal or early-life EDC exposure with elevated testicular GCT (TGCT) incidence, though causation remains correlative due to confounding variables like . Pesticides, solvents, and occupational exposures to fuels or metals also elevate GCT risk, particularly TGCT, with studies showing increased odds ratios for maternal exposure during (e.g., petroleum solvents in older offspring). Extreme temperatures (>80°F or <60°F) in occupational settings may heighten vulnerability due to the testes' external position, exacerbating DNA damage in spermatogonia. Air pollution and radiation contribute to germ cell DNA mutations, impairing repair mechanisms and spermatogenic efficiency, as observed in cohort studies linking urban particulate exposure to reduced sperm quality. Among lifestyle factors, cigarette smoking impairs spermatogenesis by reducing sperm concentration, motility, and viability, while inducing DNA fragmentation via oxidative stress; meta-analyses report 20–30% declines in semen parameters among smokers compared to non-smokers. Heavy alcohol consumption (>14 units/week) similarly elevates sperm DNA fragmentation index (SDFI) and compromises , with dose-dependent effects documented in semen analyses of chronic drinkers. , defined by >30 kg/m², disrupts germ cell function through hypothalamic-pituitary-gonadal axis alterations, increased conversion, and pro-inflammatory cytokines that promote in spermatocytes and oocytes; longitudinal data indicate 10–15% lower fertilization rates in obese males. High-fat diets and sedentary behavior exacerbate these effects by impairing energy metabolism in germ cells, though interventions like can partially restore parameters.

Induced Differentiation and Therapeutic Applications

In Vitro Gametogenesis from Stem Cells

In vitro gametogenesis (IVG) refers to the process of generating functional gametes—spermatozoa or oocytes—from pluripotent stem cells (PSCs), such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs), by recapitulating key stages of germ cell development outside the body. This approach begins with somatic cells into iPSCs, followed by directed into primordial germ cell-like cells (PGCLCs), induction of , and maturation into haploid gametes. IVG holds potential for treating , preserving in cancer patients, and enabling same-sex or single-parent , though it remains experimental in humans.00144-9) Pioneering work in mice demonstrated the feasibility of complete IVG cycles as early as 2012, where iPSCs were differentiated into functional oocytes or capable of producing viable upon fertilization and transfer. In these protocols, PGCLCs are co-cultured with gonadal cells to form gonad-like structures, promoting epigenetic erasure, sex-specific , and meiotic progression. By 2023, refinements included three-dimensional culture systems that yielded high-quality oocytes from premeiotic germ cells, surpassing traditional two-dimensional methods in efficiency and cost-effectiveness. Similar successes extended to , with iPSC-derived achieving fertilization rates comparable to natural gametes in mouse models. In humans, IVG has advanced more incrementally due to ethical constraints and technical hurdles like incomplete epigenetic and low meiotic fidelity. A 2024 study reconstituted epigenetic resetting in germ cells from PSCs, mimicking the erasure of imprints essential for totipotency. By August 2025, researchers initiated under defined conditions from iPSCs, generating meiotic cells with proper and recombination markers, though full maturation remains elusive. A notable breakthrough in September 2025 involved deriving 82 functional oocytes from skin cell-derived iPSCs at ; these were fertilized via fertilization (IVF), yielding embryos, though long-term viability and safety require further validation. Progress in non- primates has paralleled efforts, with partial achieved but no complete cycles reported. Key challenges include ensuring genomic integrity during meiosis, avoiding aneuploidy from faulty recombination, and verifying imprinting fidelity to prevent developmental disorders in offspring. Efficiencies remain low—often below 10% for human PGCLC induction—and scalability for clinical use is limited. While mouse models provide proof-of-principle, interspecies differences in germ cell specification necessitate human-specific optimizations. Ongoing research emphasizes reducing mutation rates through enhanced DNA repair mechanisms in cultured cells, with 2025 studies exploring cell division controls to minimize errors in iPSC-derived gametes. Ethical debates center on safety risks, such as off-target genetic changes, and societal implications, prompting updated guidelines from bodies like the International Society for Stem Cell Research.00118-3)

Clinical Trials and Recent Advances (2023–2025)

As of 2025, no clinical trials have commenced for full (IVG) involving the complete derivation of functional gametes from induced pluripotent cells (iPSCs) without reliance on donor gametes, due to persistent technical, ethical, and regulatory barriers. Preclinical research has advanced, however, with key milestones including a May 2024 study from demonstrating epigenetic reprogramming of human primordial germ cell-like cells (hPGCLCs) into mitotic pro-spermatogonia and oogonia via (BMP) signaling, achieving over 10 billion-fold amplification while mimicking natural profiles. This work, published in , represents a step toward competent gametes but remains limited to early-stage cells unsuitable for fertilization. In September 2025, researchers at reported generating 82 functional human eggs from adult skin cells using a mitomeiosis-inducing cloning technique akin to , though 91% of resulting embryos exhibited genetic abnormalities precluding implantation. These eggs reached the stage at a 9% rate post-fertilization, highlighting potential for treatments in women or same-sex couples but underscoring needs for chromosomal stability improvements before clinical translation. The nearest therapeutic application in trials involves Gameto's Fertilo platform, which employs iPSC-derived ovarian support cells to enable maturation of immature eggs retrieved via minimal hormonal stimulation, entering Phase 3 clinical evaluation in January 2025 after FDA clearance. This randomized, double-blind trial assesses egg maturation efficacy, yield, and safety, aiming to reduce hormone doses by up to 80% compared to standard IVF; early showed doubled rates versus conventional maturation, with the first live birth reported in May 2025. While not germ cell generation, Fertilo exemplifies cell-induced for enhancement, bridging preclinical IVG toward practical use amid ongoing debates on editing risks.

Scientific Challenges and Ethical Controversies

A primary scientific challenge in (IVG) lies in the low efficiency of differentiating induced pluripotent cells (iPSCs) into cell-like cells (PGCLCs), with protocols often achieving yields below 10% under optimized conditions, far short of the scalability required for clinical applications. Epigenetic reprogramming represents another hurdle, as in vitro processes frequently fail to fully erase and re-establish genomic imprints, resulting in retained patterns—such as those mediated by UHRF1 in humans—that differ from development and risk imprinting disorders in resulting embryos. Inducing poses particular difficulties, especially in , where mammalian models beyond mice have not progressed beyond meiotic I, with systems showing asynchronous timing spanning weeks rather than days and elevated rates due to incomplete and recombination. Safety validation remains unresolved, with concerns over genomic instability, including off-target mutations from reprogramming and differentiation, alongside tumorigenic potential from incompletely differentiated iPSC remnants that could persist in gametes. Functional maturity of IVG-derived gametes is unproven in humans; while mouse oocytes have yielded live offspring at rates of 0.3–3.9%, human equivalents, such as those generated from skin cell-derived iPSCs in 2025 experiments, have not yet demonstrated full fertility or long-term offspring viability without xenogeneic support. Species-specific differences exacerbate these issues, as human germ cell specification relies on distinct transcription factors like SOX17 rather than the BMP4-PRDM1 axis dominant in mice, necessitating tailored culture systems that recapitulate gonadal niches absent in current two-dimensional or organoid models. Ethically, IVG prompts debates over , as somatic cells from donors—such as biopsies—could be reprogrammed into s without explicit reproductive , raising risks of non-consensual parentage akin to unauthorized . It facilitates unconventional reproduction, including genetically related offspring for same-sex couples via cross-sex derivation (e.g., gametes from iPSCs), which challenges legal definitions of parentage and while potentially commodifying genetic contributions. Equity concerns are acute, given IVG's projected high costs and technical demands, which could restrict access to wealthy individuals and perpetuate reproductive inequalities, as evidenced by surveys highlighting fears of elite-only technologies. Regulatory gaps compound these issues, with bodies like the FDA requiring unprecedented preclinical data on multigenerational safety before approving clinical IVG, amid prohibitions on human embryo creation for in jurisdictions such as the . Critics argue that while no inherent moral bar exists to IVG for treatment, empirical safety thresholds—demanding equivalence to IVF's decades of data—must precede use, lest unverified risks impose heritable harms on . Broader controversies include potential misuse for eugenic selection or solitary , underscoring the need for frameworks prioritizing empirical validation over speculative benefits.

References

  1. [1]
    Definition of germ cell - NCI Dictionary of Cancer Terms
    (jerm sel) A cell that develops into a reproductive cell, which is an egg in females and a sperm in males.
  2. [2]
    Two types of germ cells, the sexual reproduction cycle, and the ... - NIH
    May 2, 2017 · However, “germ cell” specifically denotes a progenitor cell that culminates in a gamete, or the sexual reproductive cell at any stage from the ...
  3. [3]
    Mammalian primordial germ cell specification - PMC - PubMed Central
    This Review discusses the induction of PGCs in key mammalian models, including the new stem cell-based embryo models.
  4. [4]
    Primordial Germ Cells and Sex Determination in Mammals - NCBI
    These primordial germ cells migrate to the developing gonads, which will form the ovaries in females and the testes in males. After a period of mitotic ...
  5. [5]
    Primordial Germ Cell Development - UNSW Embryology
    Feb 26, 2022 · Primordial Germ Cells (PGCs) are thought to be the first population of cells to migrate through the primitive streak in early gastrulation.Introduction · Some Recent Findings · Primordial Germ Cell Migration · Molecular
  6. [6]
    primordial germ cell migration through the developing mouse embryo
    Jul 17, 2025 · The migration of primordial germ cells (PGCs), which eventually give rise to sperm and egg, is essential for fertility and evolutionarily ...
  7. [7]
    Germ Line - National Human Genome Research Institute
    Egg and sperm cells are called germ cells, in contrast to the other cells of the body, which are called somatic cells.
  8. [8]
    Primordial Germ Cells: Current Knowledge and Perspectives - PMC
    The primordial germ cell (PGC) is the primary undifferentiated stem cell type that will differentiate towards gametes: spermatozoa or oocytes.
  9. [9]
    Review Fetal germ cell development in humans, a link with infertility
    We will discuss the main events during human fetal (female) early gametogenesis and how the dysregulation of this highly complex and lengthy process can link ...
  10. [10]
  11. [11]
    Germline stem cells in human | Signal Transduction and Targeted ...
    Oct 2, 2022 · The germline cells are essential for the propagation of human beings, thus essential for the survival of mankind. The germline stem cells, ...
  12. [12]
  13. [13]
    Somatic Cells - National Human Genome Research Institute
    Somatic cells are the cells in the body other than sperm and egg cells (which are called germ cells). In humans, somatic cells are diploid.
  14. [14]
    The deteriorating soma and the indispensable germline: gamete ...
    Dec 18, 2019 · However, recent research has revealed that the so-called 'Weismann Barrier' is leaky, and that information is transferred from soma to germline.
  15. [15]
    The immortality of germ cells - Whitehead Institute - MIT
    Oct 31, 2022 · Early in embryonic development, the germline is set aside from the cells that make up the rest of the body, which are called somatic cells. The ...
  16. [16]
    Germ cells, origin of somatic stem cells? | Cell Research - Nature
    Aug 4, 2008 · Germ cells are highly specialized cells that form gametes, and they are the only cells within an organism that contribute genes to offspring ...
  17. [17]
    Somatic Cell - an overview | ScienceDirect Topics
    Somatic Cells Divide by Mitosis; Germ Cells Divide by Meiosis. Humans have in general two types of cells: the somatic cells make up the vast majority of the ...
  18. [18]
    Molecular Regulation of the Mitosis/Meiosis Decision in Multicellular ...
    ... germ cells in a totipotent state during meiotic progression. ... Plants do not have germline stem cells, per se, but instead possess totipotent somatic cells that ...
  19. [19]
    Epigenetic regulation of germ cell differentiation - PMC - NIH
    Germ cells and somatic cells have the identical genome. However, unlike the mortal fate of somatic cells, germ cells have the unique ability to differentiate ...
  20. [20]
    Pathway to Totipotency: Lessons from Germ Cells - ScienceDirect.com
    Dec 1, 2006 · In this Review, we discuss characteristics that distinguish germ cells from somatic cells and speculate on the mechanisms that allow germ cells to develop, ...
  21. [21]
    Breaking the germ line–soma barrier - Nature
    Feb 10, 2016 · This demarcation between the germ line and the soma is apparently never breached, which is known as the Weismann barrier. Indeed, under normal ...
  22. [22]
    Bubbling beyond the barrier: exosomal RNA as a vehicle for soma ...
    Nov 7, 2023 · 'Weismann's barrier' has restricted theories of heredity to the transmission of genomic variation for the better part of a century.<|control11|><|separator|>
  23. [23]
    Evolving Separate Tasks — How Somatic and Germ Cells Parted ...
    the day-to-day work of keeping a body alive — while the germ cells remain above the fray, waiting ...
  24. [24]
    Germ Line Versus Soma in the Transition from Egg to Embryo
    The germ line retains reproductive potential, while the soma are non-reproductive cells. The timing of their separation varies, and the germ line may acquire ...
  25. [25]
    A pan-metazoan view of germline-soma distinction challenges our ...
    The essential feature of germline-soma distinction is the continual presence of both germline cells that can pass on genetic information to the next generation ...
  26. [26]
    Causes and evolutionary consequences of primordial germ-cell ...
    Jun 5, 2017 · Primordial germ cells (PGCs) are specialized cells located in sexual organs of animals. These cells are central to reproduction because they ...<|control11|><|separator|>
  27. [27]
    Contrasting patterns of molecular evolution in metazoan germ line ...
    Feb 11, 2019 · The germ lines first arise from primordial germ cells (PGCs) during embryogenesis: these form from either a presumed derived mode of preformed ...
  28. [28]
    Mechanisms of germ cell specification across the metazoans
    Dec 15, 2003 · Germ cells play a unique role in gamete production, heredity and evolution. Therefore, to understand the mechanisms that specify germ cells ...
  29. [29]
    Evolving Lessons on Metazoan Primordial Germ Cells in Diversity ...
    May 11, 2025 · Primordial germ cells (PGCs) are the precursor/progenitor of the metazoan germline. The PGCs originate from developing embryonic tissues and ...
  30. [30]
    Evolution of germline segregation processes in animal development
    Apr 10, 2015 · Germline segregation is a complex process by which germline cells are separated from somatic tissues during development.Introduction · Other modes of germline... · Evolutionary scenarios for...
  31. [31]
    August Friedrich Leopold Weismann (1834-1914)
    May 23, 2014 · Weismann proposed the theory of the continuity of germ-plasm, a theory of heredity. Weismann ... "August Weismann's Theory of the Germ-Plasm and ...
  32. [32]
    Environmentally Induced Epigenetic Transgenerational Inheritance ...
    Dec 4, 2020 · For the past 120 years, the Weismann barrier and associated germ plasm theory of heredity have been a doctrine that has impacted ...
  33. [33]
    The germ-plasm; a theory of heredity : Weismann, August, 1834-1914
    Feb 11, 2009 · The germ-plasm; a theory of heredity. xxiii, 477 p. 19 cm. Show More Show Less. This book is available with additional data at Biodiversity Heritage Library.
  34. [34]
    Avian Primordial Germ Cell Migration - PubMed
    Jul 31, 2025 · Since Swift's discovery of primordial germ cells (PGCs) within the vasculature of chicken embryos in 1914, significant progress has been ...
  35. [35]
    Transplantation of Posterior Polar Plasm in Drosophila. - PNAS
    Since single cells can also be transplanted, it should be possible to discover the developmental fate of individual blastoderm cells. In recent experiments 20% ...
  36. [36]
    A germ cell origin of embryonic stem cells? | Development
    Jan 15, 2005 · In elegant, clonal-fate mapping studies in the mouse(Lawson and Hage, 1994), germ cells were shown to arise from a founder population in the E6.<|separator|>
  37. [37]
    Germ Cell Specification - PMC - NIH
    Germ cell specification in C. elegans involves the germ plasm, inherited from P4, which inhibits mRNA transcription, regulates translation, and establishes ...
  38. [38]
    Primordial Germ Cell Specification in Vertebrate Embryos
    Sep 16, 2021 · Here, we review the two main mechanisms of PGC specification, induction, and preformation, in the context of four model vertebrate species.
  39. [39]
    [PDF] Mechanisms of germ cell specification across the metazoans
    In the following sections, we review data on the earliest specification of germ cells in development, in both the. Germ cells play a unique role in gamete ...<|separator|>
  40. [40]
    Mammalian primordial germ cell specification | Development
    Mar 15, 2021 · In the mouse, a tissue called the extra-embryonic ectoderm (ExE) is primarily responsible for providing the crucial signals that induce mPGC ...
  41. [41]
    Preformation and epigenesis converge to specify primordial germ ...
    Jan 5, 2022 · In epigenesis, PGC specification is non-autonomous and depends on extrinsic signaling pathways. The BMP pathway provides the key PGC ...
  42. [42]
    Structural and functional organization of germ plasm condensates
    In C. elegans, the germ granules and the surrounding germ plasm are maternally loaded into the oocyte. After fertilization and through four asymmetric ...
  43. [43]
    Germ Plasm Assembly and Germ Cell Migration in Drosophila
    In Drosophila, germ plasm is assembled during oogenesis at the posterior pole of the growing oocyte. Germ plasm contains the posterior determinant nanos (nos), ...
  44. [44]
    Origin and establishment of the germline in Drosophila melanogaster
    Apr 4, 2025 · Electron microscope studies by Mahowald revealed the fine structure of polar granules in the germplasm of Drosophila embryos (also called germ ...
  45. [45]
    Germ plasm in <i>Caenorhabditis elegans, Drosophila</i> and <i ...
    Thus, the P granules are always inherited into germline cells, or germ cells, at all stages of the life cycle in embryos, larvae, and adults (Strome & Wood 1982) ...
  46. [46]
    Germ plasm in Caenorhabditis elegans, Drosophila and Xenopus
    Special cytoplasm, called germ plasm, that is essential for the differentiation of germ cells is localized in a particular region of Caenorhabditis elegans, ...Missing: inheritance | Show results with:inheritance
  47. [47]
    Caenorhabditis elegans germ granules accumulate hundreds of low ...
    Jul 10, 2024 · In animals with germ plasm, embryonic germline precursors inherit germ granules, condensates proposed to regulate mRNAs coding for germ cell ...INTRODUCTION · RESULTS · DISCUSSION · MATERIALS AND METHODS
  48. [48]
    Evolution of the Germ Line - Extavour Lab
    Animal germ cells can be specified during embryogenesis by inheriting maternal determinants present in the egg, by receiving inductive signals from ...
  49. [49]
    [PDF] Convergent evolution of germ granule nucleators - Extavour Lab
    Recent ad- vances in our understanding of animals that inherit germ granules sug- gest that germ plasm and germ granules are not equivalent, and may in fact ...
  50. [50]
    Primordial Germ Cell Specification in Vertebrate Embryos - Frontiers
    Here, we review the two main mechanisms of PGC specification, induction, and preformation, in the context of four model vertebrate species.
  51. [51]
    Mechanisms guiding primordial germ cell migration - PubMed Central
    Primordial germ cells (PGCs), precursors to sperm and eggs, have to migrate across the embryo to reach somatic gonadal precursors (SGPs) and fulfill their ...
  52. [52]
    Mechanisms guiding primordial germ cell migration: strategies from ...
    PGC migration in all species follows similar steps: initiation of polarity and directed migration, regulated migration by attractive and repulsive cues, and ...
  53. [53]
    advances and promises in the study of primordial germ cell migration
    Apr 12, 2024 · Primordial germ cells (PGCs) migrate through many tissues to reach the gonad, responding to cues and often starting at the periphery of the ...
  54. [54]
    advances and promises in the study of primordial germ cell migration
    This Review covers the mechanisms of primordial germ cell migration in model organisms and systems, emphasizing the guidance factors and signaling pathways.
  55. [55]
    Guidance of primordial germ cell migration by the chemokine SDF-1
    Knocking down SDF-1 or its receptor CXCR4 results in severe defects in PGC migration. Specifically, PGCs that do not receive the SDF-1 signal exhibit lack of ...
  56. [56]
    Germ Cell Migration - Developmental Biology - NCBI Bookshelf
    Unlike those of amphibians and mammals, the PGCs of birds and reptiles migrate to the gonads primarily by means of the bloodstream (Figure 19.11). When ...
  57. [57]
    Impaired colonization of the gonads by primordial germ cells in mice ...
    Here, we show that SDF-1 is not required for directed migration through tissues of embryos but instead is essential for the homing of PGCs into the genital ...
  58. [58]
    Epigenetic reprogramming in mouse and human primordial germ cells
    Dec 13, 2024 · Primordial germ cells (PGCs) are the precursors of sperm and eggs. They undergo genome-wide epigenetic reprogramming to erase epigenetic memory and reset the ...
  59. [59]
    Global profiling of DNA methylation erasure in mouse primordial ...
    We show that DNA methylation erasure is global and affects genes of various biological functions. We also reveal complex kinetics of demethylation that are ...
  60. [60]
    Primordial germ cell DNA demethylation and development require ...
    May 3, 2024 · The expression of germ cell specific genes in PGCs is activated during epigenetic reprogramming, specifically through DNA demethylation of gene ...
  61. [61]
    Epigenetic reprogramming enables the primordial germ cell-to ...
    Sep 7, 2018 · 5 Tet1-KO PGCs, our study clearly documents that Tet1 stimulates transcription of GRR genes also via a DNA demethylation-independent mechanism,.
  62. [62]
    DNA methylation dynamics during epigenetic reprogramming in the ...
    DNA methylation poses a fundamental epigenetic barrier that guides and restricts differentiation and prevents regression into an undifferentiated state.
  63. [63]
    Epigenetic resetting in the human germ line entails histone ...
    Jan 18, 2023 · Epigenetic resetting in the mammalian germ line entails acute DNA demethylation, which lays the foundation for gametogenesis, totipotency, and embryonic ...<|separator|>
  64. [64]
    Erasure of DNA methylation, genomic imprints, and epimutations in ...
    Aug 2, 2016 · We have shown that an in vitro cell culture model of mouse primordial germ cells effectively recapitulates the process of germline epigenetic erasure.
  65. [65]
    Erasure of DNA methylation, genomic imprints, and epimutations in ...
    We have shown that an in vitro cell culture model of mouse primordial germ cells effectively recapitulates the process of germline epigenetic erasure.
  66. [66]
    Reinforcement of repressive marks in the chicken primordial germ ...
    Apr 26, 2024 · This process represents an epigenome reprogramming, erasing inherited epigenetic information from gametes and preimplantation development before ...
  67. [67]
    Genomic imprinting in mammals: its life cycle, molecular ...
    The paternal and maternal epigenetic imprints established in the germline are transmitted to the zygote through fertilization and maintained faithfully ...
  68. [68]
    New insights into establishment and maintenance of DNA ...
    Fundamental to genomic imprinting in mammals is the acquisition of epigenetic marks that differ in male and female gametes at 'imprinting control regions' ...
  69. [69]
    The specification of imprints in mammals | Heredity - Nature
    Jun 18, 2014 · At the heart of genomic imprinting in mammals are imprinting control regions (ICRs), which are the discrete genetic elements that confer imprinted monoallelic ...
  70. [70]
    Maternal Primary Imprinting Is Established at a Specific Time for ...
    Primary imprinting during gametogenesis governs the monoallelic expression/repression of imprinted genes in embryogenesis. Previously, we showed that ...
  71. [71]
    Genomic imprinting: employing and avoiding epigenetic processes
    Once imprints are established in the germline, it is imperative that they survive both the reprogramming that occurs in the preimplantation embryo and the ...
  72. [72]
    The role and mechanisms of DNA methylation in the oocyte
    Nov 29, 2019 · Methylation increases during oocyte growth and is completed by the FGO GV stage. High transcriptional levels, DNA methylation and H3K36me3 ...Oocyte Methylation Landscape · Dna Methylation Machinery · Recruitment Of Dnmts In The...Missing: spermatocytes | Show results with:spermatocytes
  73. [73]
    Genomic imprinting in germ cells: imprints are under control in
    Imprint establishment starts before birth, in pro-spermatogonia (at around 14.5 dpc), is completed in peri-natal pro-spermatogonia, and is maintained through ...
  74. [74]
    Genomic imprinting in development, growth, behavior and stem cells
    May 1, 2014 · Imprints are acquired in a sex-specific manner in the mature germline (light green circles) during development, with paternal imprints (blue ...
  75. [75]
    Genomic Imprinting During Seed Development - ScienceDirect.com
    Genomic imprinting allows parent-of-origin specific control over gene expression. Although imprinted genes (or entire chromosomes) are homologous sequences ...
  76. [76]
    Meiosis - Developmental Biology - NCBI Bookshelf - NIH
    After the germ cell's last mitotic division, a period of DNA synthesis occurs, so that the cell initiating meiosis doubles the amount of DNA in its nucleus. In ...
  77. [77]
    The Beginning of Meiosis in Mammalian Female Germ Cells
    Oct 20, 2022 · Meiosis is the unique division of germ cells resulting in the recombination of the maternal and paternal genomes and the production of haploid gametes.
  78. [78]
    Genetics, Meiosis - StatPearls - NCBI Bookshelf - NIH
    In egg cells, the female gametes, meiosis follows the same general phases with only a slight variation. During telophase I, the cytoplasm divides unequally ...
  79. [79]
    Cell cycle regulation for meiosis in mammalian germ cells - PMC - NIH
    This review discusses germ cell development from the viewpoint of cell cycle regulation and highlights the mechanism of the entry of germ cells into meiosis.
  80. [80]
    Mechanism of initiation of meiosis in mouse germ cells - PubMed
    Jun 3, 2022 · Meiosis is critical for germ cell development in multicellular organisms. Initiation of meiosis coincides with pre-meiotic S phase, ...
  81. [81]
    Retinoic acid, meiosis and germ cell fate in mammals | Development
    Oct 1, 2007 · During embryogenesis, germ cells in an ovary enter meiosis, thereby committing to oogenesis. By contrast, germ cells in a testicular environment ...
  82. [82]
    Male Germ Cell Specification and Differentiation - ScienceDirect.com
    Primordial germ cell (PGC) specification or formation marks the initiation of the life cycle of germ cell lineage in all species. Although no obvious sexual ...
  83. [83]
    Control of mammalian germ cell entry into meiosis - ScienceDirect.com
    Germ cells enter meiosis at a different time between males and females. A complex suite of molecular signals acts co-ordinately to ensure correct sex-specific ...Missing: adaptations | Show results with:adaptations
  84. [84]
    Meiosis and beyond – understanding the mechanistic and ...
    ... differences observed between male and female meiosis. (1). Genome organisation in male meiosis. In mouse spermatocytes during prophase, telomeres are ...
  85. [85]
    Not all germ cells are created equal: Aspects of sexual dimorphism ...
    In meiosis II, sister chromatids are separated into daughter cells creating, in males at least, four haploid cells (or gametes). In females, half of the ...
  86. [86]
    The role of sex chromosomes in mammalian germ cell differentiation
    Hence, in normal mammalian development, male germ cells differentiate in the presence of X and Y chromosomes, and female germ cells do so in the presence of two ...
  87. [87]
    Meiosis, Genetic Recombination, and Sexual Reproduction - Nature
    In mammals, the number of viable gametes obtained from meiosis differs between males and females. In males, four haploid spermatids of similar size are produced ...
  88. [88]
    Differential susceptibility of male and female germ cells to ... - eLife
    Jan 16, 2024 · Female germ cells are resistant to glucocorticoid signaling, while male germ cells are susceptible, showing a sexual dimorphism in GR function.
  89. [89]
    Leagues of their own: sexually dimorphic features of meiotic ...
    Mar 2, 2019 · However, a growing body of evidence indicates that meiotic mechanisms differ between oogenesis and spermatogenesis. One of the largest ...
  90. [90]
    From primordial germ cells to primordial follicles: a review and visual ...
    Jun 21, 2016 · The major developmental events were divided into five phases: 1) origin of germ cells and maintenance of pluripotency; 2) primordial germ cell ...
  91. [91]
    Prefertilization Events
    Primordial germ cells migrate into the ovaries at week 4 of development and differentiate into oogonia (46,2N). Oogonia enter meiosis I and undergo DNA ...
  92. [92]
    [PDF] Gonadogenesis, Female - skinner@wsu.edu
    when primordial germ cells (i.e., oogonia) stop pro- liferating and enter meiotic prophase. This process occurs during Embryonic Weeks 7 and 8 in humans and ...
  93. [93]
    Oogenesis - Developmental Biology - NCBI Bookshelf - NIH
    When the primary oocyte divides, its nucleus, called the germinal vesicle, breaks down, and the metaphase spindle migrates to the periphery of the cell. At ...
  94. [94]
    [PDF] The primordial pool of follicles and nest breakdown in mammalian ...
    Primordial follicles form from germ cell nests via germ cell apoptosis and somatic cell migration. Nests break down into follicles, with many nuclei lost.
  95. [95]
    Oogenesis - an overview | ScienceDirect Topics
    Oogonia originating from primordial germ cells proliferate by mitosis and form primary oocytes that arrest at the prophase of the first meiotic division.
  96. [96]
  97. [97]
    Portrait of an oocyte: our obscure origin - JCI
    The cytoplasm contains a honeycomb of ER, and the widely distributed polyribosomes increase four-fold during oocyte growth to meet the demands of protein ...
  98. [98]
    Oocyte maturation: gamete-somatic cells interactions, meiotic ...
    The oocyte produces and accumulates molecules and organelles that are fundamental for the development of the preimplantation embryo.
  99. [99]
    The molecular regulatory mechanisms of meiotic arrest and ... - NIH
    Oct 2, 2023 · The progress of the cellular and molecular mechanisms of oocyte nuclear maturation including meiosis arrest and meiosis resumption is summarized.
  100. [100]
    The art of oocyte meiotic arrest regulation - PMC - PubMed Central
    Jan 5, 2019 · This review will discuss how these two cyclic nucleotides regulate oocyte maturation by blocking or initiating meiotic processes.Intra-Oocyte Elevated Camp... · Fig. 1 · Bidirectional Communication...
  101. [101]
    Molecular determinants of the meiotic arrests in mammalian oocytes ...
    Mammalian oocytes undergo two rounds of developmental arrest during maturation: at the diplotene of the first meiotic prophase and metaphase of the second ...
  102. [102]
    [PDF] REPRODUCTION - UConn Health
    Oocytes are arrested at the first meiotic prophase, held in meiotic arrest by the surrounding follicle cells until a surge of LH from the pituitary stimulates ...
  103. [103]
    Luteinizing Hormone Action in Human Oocyte Maturation and Quality
    In women, the mid-cycle LH surge triggers GVBD, cumulus cell expansion, and extrusion of the first polar body at 15, 22, and 35 h after the start of the LH ...
  104. [104]
    Oocytes can efficiently repair DNA double-strand breaks to ... - PNAS
    May 7, 2020 · We show that when apoptosis is inhibited, oocytes can repair severe DNA damage via homologous recombination repair. The repair is highly efficient.
  105. [105]
    Distinct characteristics of the DNA damage response in mammalian ...
    Feb 14, 2024 · DSBs are repaired primarily through two major mechanisms: nonhomologous end joining (NHEJ) and homologous recombination (HR). NHEJ begins with ...
  106. [106]
    Molecular regulation of DNA damage and repair in female infertility
    Aug 14, 2024 · In this review, we summarize recent advances in the DNA damage response mechanisms in infertility diseases such as PCOS, endometriosis, diminished ovarian ...
  107. [107]
    Local DNA synthesis is critical for DNA repair during oocyte maturation
    Oct 12, 2021 · During oocyte maturation, damaged DNA is repaired primarily by non-homologous end joining (NHEJ) or homologous recombination (HR). Although ...
  108. [108]
    Changes in DNA repair compartments and cohesin loss promote ...
    Oct 21, 2024 · We show that aged oocytes have a lower DNA repair capacity and reduced mobility of DNA damage sites compared to young oocytes.
  109. [109]
    Increased DNA damage in full-grown oocytes is correlated ... - Nature
    Nov 1, 2024 · Autophagy has both DNA damage protection and repair roles in oocytes by promoting RAD51 recruitment to the DNA. To understand how autophagy ...
  110. [110]
    importance of DNA repair for maintaining oocyte quality in response ...
    Primordial follicle oocytes can efficiently repair DNA DSBs arising from diverse origins, but this capacity may decline with increasing age.
  111. [111]
    Timing, rates and spectra of human germline mutation - PMC - NIH
    3.8% of mutations were mosaic in the parental germline, resulting in 1.3% of mutations being shared between siblings. The number of these shared mutations ...
  112. [112]
    Allele frequency selection and no age-related increase in human ...
    Aug 6, 2025 · Here, using highly accurate duplex sequencing, we detected de novo mutations in single oocytes, blood, and saliva in women 20 to 42 years of age ...
  113. [113]
    A paternal bias in germline mutation is widespread in amniotes and ...
    Aug 2, 2022 · This paper challenges a fundamental view concerning why males of most animals have a higher germline mutation rate than females.
  114. [114]
    Replication Errors Made During Oogenesis Lead to Detectable De ...
    For most oocytes, this mutation rate was in the range of 10−6 mutations per nt, while oocytes 4.4, 5.10, and 7.3 had a higher mutation rate. On average, the ...
  115. [115]
    Mechanisms underlying low mutation rates in mammalian oocytes ...
    Aug 11, 2025 · Mammalian oocytes and embryos are known to exhibit a markedly low frequency of de novo mutations compared to somatic cells.
  116. [116]
    Histology, Spermatogenesis - StatPearls - NCBI Bookshelf - NIH
    The final stage of spermatogenesis includes spermatozoa production, mature and motile sperm cells, from round spermatids, through a process called ...
  117. [117]
    Review: Spermatogenesis in the bull - ScienceDirect.com
    Spermatogenesis can be divided into three parts: spermatocytogenesis, meiosis and spermiogenesis. During spermatocytogenesis, germ cells engage in a cycle of ...
  118. [118]
    Spermatogenesis - Developmental Biology - NCBI Bookshelf - NIH
    The spermatogonial stages last 8 days, meiosis lasts 13 days, and spermiogenesis takes up another 13.5 days. In humans, spermatic development takes nearly twice ...
  119. [119]
    Spermatogenesis - IntechOpen
    Spermiogenesis has basically four stages [6, 10, 11]: Golgi phase, cap phase, tail formation and maturation stage. In the Golgi stage , the initially radially ...2.1 Spermatocytogenesis · 2.3 Spermiogenesis · 2.3. 1 Phases Of...
  120. [120]
    Mechanisms of spermiogenesis and spermiation and how they are ...
    The different steps, or phases, of spermiogenesis are distinguished by the morphological appearance of the developing acrosome and the changing shape of the ...
  121. [121]
    Novel Stage Classification of Human Spermatogenesis Based on ...
    B spermatogonia are first apparent in stage I, preleptotene spermatocytes are formed in stage V, leptonema starts in stage VII, and spermiation takes place at ...
  122. [122]
    Spermatogenesis in man: an estimate of its duration - PubMed
    These results indicated that one cycle of the seminiferous epithelium lasts 16 days, and the whole of spermatogenesis is estimated to consume approximately 64 ...
  123. [123]
    Spermatogenesis in humans and its affecting factors - ScienceDirect
    The entire spermatogenic process is thought to require approximately 74 days [2], [3], but a more recent study in normal men concluded that the total time to ...
  124. [124]
    Spermatogenesis: The Commitment to Meiosis - PubMed Central - NIH
    Nov 4, 2015 · The duration of spermatogenesis in humans is reportedly 74 days where one cycle of the seminiferous epithelium is 16 days. Recently, the number ...
  125. [125]
    Development of functional spermatozoa in mammalian ...
    Jul 22, 2024 · During spermiogenesis, the nucleus is condensed. One of the major processes of nuclear condensation is the replacement of histones with ...
  126. [126]
    Spermiogenesis, Stages of Seminiferous Epithelium and Variations ...
    Mar 28, 2020 · The results of the study revealed that inside seminiferous epithelium, initial steps of spermiogenesis are depicted by changes in acrosomic ...
  127. [127]
    Formation and function of the manchette and flagellum during ...
    ... (spermiogenesis), where the nucleus is remodeled by chromatin condensation, the excess cytoplasm is removed and the acrosome and sperm tail are formed.
  128. [128]
    Iqcg Is Essential for Sperm Flagellum Formation in Mice | PLOS One
    During spermiogenesis, round spermatid undergoes dramatic morphogenesis to give rise to mature spermatozoon, including the condensation and elongation of ...
  129. [129]
    Molecular changes and signaling events occurring in sperm during ...
    During epididymal maturation, sperm acquire progressive movement, the cytoplasmic droplet migrates, and changes occur in the sperm proteome, surface, and lipid ...
  130. [130]
    New insights into epididymal function in relation to sperm maturation in
    The epididymis enables sperm to move, capacitate, and bind to the egg, acquiring motility and the ability to fuse to the oocyte, essential for fertility.The epididymal sperm... · Epididymal spermatozoa... · Relationship between...
  131. [131]
    Sperm Maturation in the Epididymis - SpringerLink
    Sperm acquire motility and fertility in the epididymis, a long tubule with unique microenvironments in each segment that allow maturation.
  132. [132]
    A novel mechanism of sperm midpiece epididymal maturation and ...
    Jul 1, 2025 · We show the sperm midpiece is structurally immature upon exiting the testis and maturation continues during transit from the caput to the cauda ...
  133. [133]
    [PDF] Recent advances in the study of the structure and function of the ...
    May 17, 2024 · In general, epididymal PCs play an essential role in sperm maturation, regulation of luminal pH, and immunomodulation. Thus, it can be seen ...
  134. [134]
    Spermatogenesis, DNA damage and DNA repair mechanisms in ...
    In the male germline cells, DNA repair mechanisms include nucleotide excision repair, base excision repair, DNA mismatch repair, double strand break repair and ...
  135. [135]
    DNA double-strand break repair in male germ cells during ...
    In this review, we comprehensively introduce DSB repair pathways being used by male germ cells during spermatogenesis.
  136. [136]
    MCM9 deficiency impairs DNA damage repair during ... - Nature
    Jul 1, 2025 · MCM9 deficiency impairs DNA damage repair during spermatogenesis, leading to Sertoli cell-only syndrome in humans.
  137. [137]
    Mechanisms of oxidative stress-induced sperm dysfunction - Frontiers
    Feb 4, 2025 · Unlike somatic cells, spermatozoa have limited DNA repair mechanisms due to the replacement of histones with protamines during spermatogenesis.
  138. [138]
    Direct measurement of the male germline mutation rate in ... - Nature
    Mar 15, 2025 · Notably, ~80% of de novo mutations arise in the paternal (male) germline, and for every additional year of paternal age, offspring inherit ~1.3– ...
  139. [139]
    Paternal Age Effect Mutations and Selfish Spermatogonial Selection
    Advanced paternal age has been associated with an increased risk for spontaneous congenital disorders and common complex diseases.
  140. [140]
    Sperm sequencing reveals extensive positive selection in the male ...
    Oct 8, 2025 · In humans, positive selection of driver mutations during spermatogenesis can increase the birth prevalence of certain developmental disorders.
  141. [141]
    Spermatogenesis, DNA damage and DNA repair mechanisms in ...
    In the male germline cells, DNA repair mechanisms include nucleotide excision repair, base excision repair, DNA mismatch repair, double strand break repair and ...
  142. [142]
    Testicular germ cell tumor: a comprehensive review - PMC - NIH
    Jan 22, 2019 · Both inherited genetic factors and environmental risk factors emerge as important contributors to TGCT susceptibility. Genome-wide association ...Missing: peer- | Show results with:peer-
  143. [143]
    Cancer Stat Facts: Testicular Cancer - SEER
    Rate of New Cases and Deaths per 100,000: The rate of new cases of testicular cancer was 6.0 per 100,000 men per year. The death rate was 0.3 per 100,000 men ...Missing: etiology | Show results with:etiology
  144. [144]
    Updates in the Management of Malignant Ovarian Germ Cell Tumors
    The cause of malignant ovarian germ cell tumors is unknown, but it is likely related to both genetic and environmental factors. 11,12 Malignant ovarian germ ...
  145. [145]
    Extragonadal germ cell tumors: Not just a matter of location. A ... - NIH
    Extragonadal germ cell tumors (EGGCTs) are a heterogeneous group of tumors of neoplastic germ cells arising from extragonadal anatomical locations.
  146. [146]
    Ovarian Germ Cell Tumors: Causes & Treatment - Cleveland Clinic
    Ovarian germ cell tumors form when sex cells (germ cells) undergo changes (mutations) and form a mass. Experts continue to research why this happens, including ...
  147. [147]
    Identification of 22 susceptibility loci associated with testicular germ ...
    Jul 23, 2021 · This meta-analysis identifies 22 TGCT susceptibility loci, bringing the total to 78, which account for 44% of disease heritability.<|separator|>
  148. [148]
    Testicular cancer in 2023: Current status and recent progress
    Nov 10, 2023 · These tumors are more common in White and Hispanic Americans than in Black Americans. Rates are increasing most rapidly among Hispanic men, and ...
  149. [149]
    Germ cell loss in Klinefelter syndrome: When and why? - PubMed
    May 15, 2020 · Infertility in KS patients is caused by a massive germ cell loss, leading to azoospermia in more than 90% of the adult patients.
  150. [150]
    The Genetics of Infertility: Current Status of the Field - PMC - NIH
    It has been shown that germ cells with an extra Y chromosome from men with the 47,XYY karyotype have abnormal meiotic pairing, suggesting disrupted meiosis, ...
  151. [151]
    Genetic Causes of Male Infertility - PMC - NIH
    Our results showed that Klinefelter's syndrome and complete AZFc deletions are the most common genetic causes of azoospermia. Partial AZFc deletions as well as ...
  152. [152]
    Y chromosome azoospermia factor region microdeletions and ...
    AZF microdeletions are recognized as the most frequent structural chromosomal abnormalities and are the major cause of male infertility.
  153. [153]
    Sertoli Cell–Only Syndrome - StatPearls - NCBI Bookshelf
    Sertoli cell-only syndrome, also known as del Castillo syndrome or germ cell aplasia, is one of the most common causes of azoospermia in infertile men.
  154. [154]
    Human male infertility and its genetic causes - PMC - PubMed Central
    Genetic causes of azoospermia include chromosomal abnormalities, Y chromosome microdeletions, and specific mutations/deletions of several Y chromosome genes.
  155. [155]
    Pathogenic variations in Germ Cell Nuclear Acidic Peptidase (GCNA ...
    Aug 20, 2021 · Pathogenic variations in Germ Cell Nuclear Acidic Peptidase (GCNA) are associated with human male infertility.
  156. [156]
    Ovarian dysfunction in women with Turner syndrome - PMC
    Mar 23, 2023 · Ovarian dysfunction is one of the most common features of women with Turner syndrome. In these women, oocyte apoptosis is markedly accelerated from the early ...
  157. [157]
    Ovarian dysfunction in women with Turner syndrome - Frontiers
    Mar 22, 2023 · Ovarian dysfunction is one of the most common features of women with Turner syndrome. In these women, oocyte apoptosis is markedly accelerated from the early ...Abstract · Ovarian features of women... · Possible causes of the ovarian...
  158. [158]
    Genetic diagnosis of subfertility: the impact of meiosis and maternal ...
    Meiosis gene mutations may therefore lead to a number of clinical pathologies such as POI, insufficient oocyte maturation and low fertilisation rate.Meiosis · Paracrine Regulation Of... · Maternal-Effect Factors
  159. [159]
    Chromosomal abnormalities predisposing to infertility, testing, and ...
    Mar 19, 2021 · Meiotic and mitotic errors during gametogenesis and fetal development, respectively, can cause chromosomal abnormalities, which predispose to infertility.
  160. [160]
    Strategies to identify genetic variants causing infertility - PMC - NIH
    Several known causative genetic aberrations are used for clinical diagnosis of male infertility, such as Klinefelter Syndrome (XXY) and Y chromosome ...
  161. [161]
    Endocrine disrupters, microRNAs, and primordial germ cells - PubMed
    Sep 15, 2016 · Endocrine-disrupting chemicals (EDCs) are environmental pollutants that may change the homeostasis of the endocrine system, altering the differentiation of ...Missing: impact | Show results with:impact
  162. [162]
    Endocrine disrupting chemicals and male fertility - Frontiers
    Endocrine disrupting chemicals (EDCs) exposure may affect male fertility at multiple levels, from sperm production and quality to the morphology and histology ...
  163. [163]
    Transgenerational Effects of Endocrine-Disrupting Chemicals on ...
    Apr 18, 2019 · Moreover, BPA disrupted germ cell nest breakdown (0.5, 20, and 50 µg/kg/d) and severely reduced fertility (0.5 µg/kg/d) in female mice (69). In ...Abstract · Phthalates · Pesticides<|separator|>
  164. [164]
    Understanding the role of endocrine disrupting chemicals in ...
    This comprehensive review examines the complex interplay between endocrine disrupting chemicals (EDCs) and the development of testicular germ cell tumors ...
  165. [165]
    Parental occupational exposure to solvents and risk of developing ...
    Jul 5, 2023 · The association found with maternal occupational exposure to fuels and petroleum solvents among older men needs further investigation. This ...
  166. [166]
    Environmental exposure to residential pesticides, chemicals, dusts ...
    Exposure to pesticides, chemicals, dusts, fumes, and unfinished metals have received considerable attention as potential risk factors for childhood GCT over the ...
  167. [167]
    Environmental toxicology of testicular cancer - ScienceDirect.com
    As the testes are outside of the body, they may be more vulnerable to environmental temperatures. Occupational exposure to extreme conditions (>80°F or <60°F) ...
  168. [168]
    How Environmental Exposures Affect Genes and Increase Cancer ...
    Mar 11, 2025 · Polluted air, radiation, tobacco smoke, and processed foods can lead to DNA damage, interfering with the body's natural ability to repair itself ...
  169. [169]
    Do Cigarette Smoking and Obesity Affect Semen Abnormality ... - NIH
    However, obesity did not affect the semen abnormality. Smoking affected semen quality and is therefore expected to play a negative role in conception.Missing: germ | Show results with:germ
  170. [170]
    Decrease in Sperm Parameters in the 21st Century: Obesity ... - MDPI
    Smoking appears to reduce sperm concentration, motility, viability, and normal morphology; also, it is associated with DNA damage and leads to the generation of ...
  171. [171]
    Impact of heavy alcohol consumption and cigarette smoking on ...
    Apr 28, 2022 · The purposes of the presents study were to investigate the impact of alcohol consumption and cigarette smoking on semen parameters and sperm DNA quality.
  172. [172]
    Impact of obesity on male fertility, sperm function and molecular ...
    There is emerging evidence that male obesity negatively impacts fertility through changes to hormone levels, as well as direct changes to sperm function and ...
  173. [173]
    Obesity and male infertility: multifaceted reproductive disruption
    Apr 4, 2022 · Increases in pro-inflammatory cytokine levels can impair male fertility via inducing germ cell apoptosis, disrupting Sertoli cell junctions, ...Obesity And Semen Parameters · Obesity And Sperm Dna... · Altered Testicular Immune...
  174. [174]
    Energy metabolism and spermatogenesis - ScienceDirect.com
    Oct 15, 2024 · Additionally, the role of lifestyle factors, such as diet, exercise, and environmental exposures, in modulating energy metabolism and ...
  175. [175]
    Stem cell-derived gametes: what to expect when expecting their ...
    Jul 2, 2025 · Overview of recent progress in the field of in vitro gametogenesis in various species. Each box shows the origin of the cells used (embryonic ...Introduction · The state-of-the-art in iPSCD... · Setting the stage for iPSCD...
  176. [176]
    In vitro oogenesis from murine premeiotic germ cells using a new ...
    Jul 31, 2023 · Compared with 2D culture and other 3D culture methods, this new culture system is more cost-effective and can create high-quality oocytes ...<|separator|>
  177. [177]
    Modelling in vitro gametogenesis using induced pluripotent stem cells
    Oct 16, 2023 · This paper extensively reviews IVG using iPSCs ... Romualdez-Tan, M.V. Modelling in vitro gametogenesis using induced pluripotent stem cells: a ...Oogenesis · Emerging Techniques · Abbreviations<|separator|>
  178. [178]
    In vitro reconstitution of epigenetic reprogramming in the human ...
    May 20, 2024 · Main. Germ cells give rise to totipotency and ensure heredity and evolution. Human PGCs are thought to be specified at around embryonic day 12 ( ...<|separator|>
  179. [179]
    Initiation of meiosis from human iPSCs under defined conditions ...
    Aug 15, 2025 · These methods for mouse in vitro gametogenesis rely on differentiating pluripotent stem cells to PGC-like cells, followed by coculture with ...
  180. [180]
    OHSU researchers develop functional eggs from human skin cells
    Sep 30, 2025 · Researchers reported that they had produced 82 functional oocytes that were then fertilized with sperm through IVF. Notably, most did not ...
  181. [181]
    Advances in in vitro oocyte generation from pluripotent stem cells ...
    Aug 10, 2025 · In human, around 24th day of gestation, PGCs form in the wall of the yolk sac and migrate to the gonadal ridges around the 5th week of gestation ...
  182. [182]
    Induction of experimental cell division to generate cells with reduced ...
    Sep 30, 2025 · Reprogramming a patient's somatic cells into induced pluripotent stem cells (iPSCs), followed by differentiation into functional sperm or ...
  183. [183]
    Groundbreaking study advances human in vitro gametogenesis for ...
    May 21, 2024 · Groundbreaking study advances human in vitro gametogenesis for infertility treatment ... Infertility affects approximately 1 in 6 people in their ...
  184. [184]
  185. [185]
    Scientists create human eggs in the lab, using skin cells - NPR
    Sep 30, 2025 · Scientists have created human eggs containing genes from adult skin cells, a step that someday could help women who are infertile or gay ...
  186. [186]
    FDA grants clearance for phase 3 trial of Fertilo, an iPSC-based ...
    Jan 30, 2025 · The phase 3 trial will evaluate the efficacy and safety of Fertilo in maturing healthy eggs outside the body, yielding embryos, and improving ...
  187. [187]
    Gameto launches Phase III iPSC fertility trial following IND greenlight
    Jan 31, 2025 · Gameto claims its Fertilo iPSC system can mature eggs outside the body using 80% fewer hormonal injections than typical methods.
  188. [188]
    The World's First Live Birth Using Fertilo - REPROCELL
    May 21, 2025 · Fertilo uses stem cells to mature eggs outside the body, reducing hormonal stimulation, side effects, and the need for costly infrastructure.
  189. [189]
    Fertilo | Gameto
    Mar 25, 2025 · Early clinical results show that Fertilo achieved a pregnancy rate more than double that of conventional in vitro maturation (IVM). This study ...
  190. [190]
    Mammalian in vitro gametogenesis | Science
    Oct 1, 2021 · Here, we will discuss the current state of mammalian in vitro gametogenesis (IVG) research (1), key challenges driving the research forward, and ...
  191. [191]
    Anticipating in vitro gametogenesis: Hopes and concerns for IVG ...
    Jun 6, 2024 · In vitro gametogenesis (IVG), the reconstitution of germ cell development in vitro, is an emerging stem cell-based technology with profound ...
  192. [192]
    In vitro gametogenesis: A review of ethical and policy questions
    Jul 24, 2025 · This policy briefing focusses on potential clinical applications of IVG in the future, and the associated ethical, legal and social issues these ...
  193. [193]
    Ethical and legal challenges in assisted same-sex conception ...
    Jan 10, 2024 · Ethical and legal challenges in assisted same-sex conception through in vitro gametogenesis. Eli Y. Adashi, Nature Medicine volume 30, pages 322–323 (2024)
  194. [194]
    Ethical and legal implications of in vitro gametogenesis and ...
    This article discusses legal and ethical issues raised by in vitro gametogenesis, including safety, Food and Drug Administration review, embryo destruction, ...
  195. [195]
    Is there a valid ethical objection to the clinical use of in vitro-derived ...
    The author concludes there is no justifiable in-principle ethical objection to IVG for those who cannot have offspring otherwise, though safety concerns exist.