Fact-checked by Grok 2 weeks ago

Spermatogonial stem cell

Spermatogonial stem cells (SSCs) are the foundational stem cells residing in the seminiferous of the testis, responsible for continuously producing spermatozoa throughout a male's reproductive lifespan by balancing self-renewal and . These diploid cells, originating from primordial germ cells during embryonic development, constitute a small fraction—approximately 0.03% (or 1 in 3,000)—of the total cells in the adult mouse testis and are defined by their unipotent capacity to regenerate the spermatogenic exclusively. SSCs are primarily located along the of the seminiferous tubules, where they interact with a specialized niche comprising Sertoli cells, peritubular myoid cells, Leydig cells, and vascular elements that provide essential regulatory signals. In , they are classified morphologically as Type A single (A_s) spermatogonia, which can divide symmetrically to maintain the pool or asymmetrically to generate progenitor cells connected by intercellular bridges, forming A_paired (A_pr) and A_aligned (A_al) configurations. This heterogeneous population exhibits varying self-renewal potentials, with subsets marked by proteins like ID4^Bright serving as the most robust stem cells capable of long-term lineage maintenance. The core biological processes governing SSCs involve cyclic regulation synchronized with the seminiferous epithelium cycle, where self-renewal is promoted by glial cell line-derived neurotrophic factor (GDNF) from Sertoli cells via the PI3K/AKT pathway, occurring roughly every 8.6 days in mice. is triggered by periodic pulses of , leading to the commitment of progenitor spermatogonia into as preleptotene spermatocytes, ultimately yielding mature in a process that produces over 10^12 gametes in a lifetime. Disruption of this balance, such as through niche alterations or genetic factors, can impair , highlighting the niche's role in SSC . Beyond their physiological role, SSCs hold significant promise in and due to their unique properties, including the ability to be isolated, cultured, and transplanted to restore in infertile models, as demonstrated in and large animal studies. Recent advances, such as serum-free culture systems and , have elucidated novel ligand-receptor interactions (e.g., PTN-SDC4) in the niche, enabling improved maintenance and genetic modification for transgenesis without ethical concerns associated with embryonic cells. As of 2025, further developments include novel gel-based systems enabling over fourfold expansion of undifferentiated spermatogonia and testicular organoids for modeling in therapeutic applications. These developments also support applications in preservation for cancer patients and through SSC banking and xenografting.

Fundamental Biology

Definition and Role in Spermatogenesis

Spermatogonial stem cells (SSCs) are a subset of type A spermatogonia in the adult testis, defined as the most primitive spermatogonia capable of both self-renewal and differentiation to sustain lifelong spermatogenesis. These cells maintain the germline stem cell pool by balancing proliferation that replenishes their own numbers with the generation of daughter cells committed to differentiation, ensuring the continuous production of spermatozoa. In mammals, SSCs reside along the basement membrane of the seminiferous tubules and function as unipotent stem cells specifically dedicated to the spermatogenic lineage. The primary role of SSCs in is to support uninterrupted production throughout reproductive life, with SSCs undergoing symmetric and asymmetric divisions, balancing self-renewal (producing two stem cells) with the generation of progenitor cells committed to . In humans, this process enables the daily generation of approximately 150–200 million spermatozoa, a scale essential for male fertility. Through clonal expansion and sequential steps, SSC-derived progenitors progress to spermatocytes, undergo to form spermatids, and ultimately mature into functional , thereby perpetuating the male germline. Developmentally, SSCs originate from fetal gonocytes, which are prospermatogonia derived from primordial germ cells that migrate to the during embryogenesis. Postnatally, these gonocytes colonize the seminiferous cords and transition into undifferentiated type A spermatogonia, establishing the SSC pool; full spermatogenic activity commences at when hormonal cues trigger their proliferation and . This transition marks the onset of continuous , with SSCs becoming the foundational elements for all subsequent production. A distinguishing feature of SSCs among is their unique capacity to transmit genetic information across generations, as they are the sole source of spermatozoa that fertilize oocytes and propagate the species. Unlike multipotent or pluripotent stem cells, which regenerate tissues, SSCs are committed exclusively to , ensuring the of genetic material without contributions. This germline-specific function underscores their irreplaceable role in and fertility.

Location in the Testis

Spermatogonial stem cells (SSCs) are located in the basal compartment of the seminiferous tubules within the testis, where they adhere directly to the . This positioning positions them below the tight junctions formed by Sertoli cells, which delineate the boundary between the basal and adluminal compartments of the seminiferous epithelium. The provides structural support and facilitates the interaction of SSCs with surrounding cells essential for their maintenance. The primary surrounding cell types include Sertoli cells, which extend from the basement membrane to the tubule lumen and provide nutritional and structural support to during . Leydig cells, situated in the interstitial space outside the tubules, produce testosterone and other hormones that influence development. Peritubular myoid cells encircle the outer surface of the seminiferous tubules, contributing contractile properties that aid in the transport of spermatozoa and testicular fluid. Within the seminiferous tubules, SSCs are organized into clusters distributed along the length of the , typically appearing as single cells (A_s) or short chains (A_pr and A_al) in . These stem cells constitute a rare population, representing approximately 0.03% of the total cells in the adult testis. During the transition from fetal to adult stages, gonocytes—precursors to SSCs—proliferate and migrate from the center of the seminiferous cords to the shortly after birth, establishing the definitive SSC location. This postnatal relocation is critical for integrating SSCs into the basal compartment and initiating continuous .

Identification and Environment

Nomenclature Across Species

The nomenclature for spermatogonial stem cells (SSCs) traces its origins to 19th-century advancements in , which first enabled the and of germ cells in the testis. The term "spermatogonium" was introduced around 1871 by Victor von Ebner to describe the undifferentiated precursors of spermatozoa observed in histological sections. Subsequent refinements in the mid-20th century led to more precise subtype classifications, particularly through radioautographic studies in . In 1971, Claire Huckins and Edgar F. Oakberg independently proposed models delineating SSC dynamics, emphasizing a of stem cells that balance self-renewal and to sustain . In mice and other , SSCs are primarily identified among undifferentiated type A spermatogonia, classified based on their configuration and intercellular connections as viewed under light microscopy. These include A_single (As) spermatogonia, which are isolated cells considered the true reservoir; A_paired (Apr) spermatogonia, formed by division of As cells; and A_aligned (Aal) spermatogonia, which form chains of 4, 8, or 16 cells connected by cytoplasmic bridges. The As subtype is functionally validated as the stem cell pool through transplantation assays, where only these cells can repopulate recipient testes and restore , producing donor-derived colonies. This nomenclature, rooted in Huckins and Oakberg's 1971 models, highlights the nature of SSC renewal, where As cells can either self-renew or commit to via Apr and Aal intermediates. In humans and nonhuman , SSC nomenclature differs due to distinct histological features and lacks the precise chain-based classification seen in . Subtypes are distinguished as A_dark (Adark) and A_pale (Apale) spermatogonia based on intensity in histological preparations, with Adark cells appearing darkly stained and Apale cells lighter. Adark spermatogonia are regarded as reserve cells with slow-cycling properties, serving as a quiescent pool activated during regeneration, while Apale spermatogonia are proliferative and renewing, actively contributing to daily spermatogenic output. Both subtypes exhibit potential, though functional confirmation remains limited compared to models. Identifying SSCs across species presents significant challenges, primarily due to the absence of universal molecular markers that definitively distinguish stem cells from progenitors. While morphological and histological criteria provide initial classification, they are insufficient for pure isolation, leading to reliance on functional assays such as transplantation or colony-forming unit tests to confirm stemness. Recent advances as of 2025, including single-cell RNA sequencing (scRNA-seq) and spectral cytometry, have refined the identification of human SSC subpopulations by revealing transcriptional states and novel marker panels, such as those delineating at least four distinct states beyond the traditional Adark/Apale hierarchy, suggesting a more dynamic model. These assays, pioneered in mice, demonstrate SSC activity retrospectively but are technically demanding in humans, underscoring ongoing needs for species-specific validation.

Stem Cell Niche

Sertoli cells form the primary structural component of the (SSC) niche, enveloping SSCs and providing physical support through intimate cell-cell contacts along the of the seminiferous tubules. These cells secrete key paracrine factors, such as glial cell line-derived neurotrophic factor (GDNF), which binds to receptors on undifferentiated spermatogonia, including SSCs, to promote their adhesion and maintenance within the niche. GDNF expression in Sertoli cells is dynamically regulated and peaks during specific stages of the seminiferous epithelial cycle, ensuring localized support for SSC self-renewal. The extracellular matrix (ECM) of the basement membrane, composed primarily of collagen IV and laminin, anchors SSCs to the niche via integrin-mediated interactions, such as α6β1-integrins binding to laminin. Collagen IV chains, including α3(IV), contribute to the structural integrity of this membrane, while laminin variants like laminin-α2 facilitate adhesion between Sertoli cells and germ cells. The blood-testis barrier (BTB), formed by tight junctions between adjacent Sertoli cells, isolates premeiotic germ cells, including SSCs, from vascular and immune components, creating a protected microenvironment that prevents immune surveillance while allowing nutrient exchange. Vascular elements near the basement membrane further support SSC localization by delivering systemic signals, though the BTB restricts direct vascular access to the adluminal compartment. Age-related alterations in the SSC niche, particularly in function, lead to reduced SSC numbers and activity, with declining sharply after 12 months in mice due to niche deterioration rather than intrinsic SSC aging. GDNF levels in s initially increase in aging-fertile males but decrease in infertile ones, contributing to niche failure. Hormonal influences, such as (FSH), enhance secretion of niche factors like GDNF and (bFGF), supporting SSC proliferation and survival through pathways including /PKA and PI3K/Akt. The SSC niche exhibits similarities across species, with conserved roles for Sertoli cells and shared ligand-receptor interactions, such as GDNF signaling, in both mice and s. However, the human niche displays greater heterogeneity, characterized by varied expression of paracrine factors like IGFBP7 and CCL24 across niche types, likely due to the extended reproductive lifespan requiring adaptive responses to prolonged demands.

Regulatory Mechanisms

Self-Renewal Pathways

Self-renewal of (SSCs) is governed by a delicate balance of extrinsic paracrine signals from the niche and intrinsic transcriptional programs that promote proliferation while preventing differentiation, ensuring the long-term maintenance of the pool. These mechanisms allow SSCs to undergo asymmetric or symmetric divisions, sustaining throughout adulthood. A primary extrinsic regulator is glial cell line-derived neurotrophic factor (GDNF), secreted by Sertoli cells, which binds to the RET/GFRα1 receptor complex on SSCs to initiate signaling cascades that drive proliferation and self-renewal. This interaction activates the PI3K-Akt pathway, enhancing survival and mitotic activity without committing cells to . Disruption of GDNF signaling, such as through receptor inhibition, reduces SSC numbers but permits recovery via compensatory self-renewing divisions upon restoration. Additional paracrine factors contribute to this process, including fibroblast growth factor 2 (FGF2) from Sertoli and endothelial cells, which promotes SSC self-renewal by activating /ERK signaling and upregulating genes like Ets variant 5 (Etv5) and B cell CLL/lymphoma 6 member B (Bcl6b). Similarly, , produced by Sertoli cells, signals through the receptor to maintain SSC homing and survival within the niche, with CXCR4-expressing cells exhibiting enriched potential. Inhibition of activity, often modulated by these signals, further balances self-renewal against ; for instance, transient suppression prevents excessive progenitor commitment, while sustained inhibition impairs SSC maintenance. Intrinsic factors, particularly transcription factors, reinforce the undifferentiated state of SSCs. Promyelocytic leukemia zinc finger (PLZF, encoded by Zbtb16) acts as a repressor of differentiation genes and opposes hyperactivity through , thereby preserving progenitor self-renewal. Inhibitor of DNA binding 4 (ID4) is selectively expressed in single As spermatogonia, marking the most primitive SSCs and regulating their transition to progenitor states by modulating levels that dictate stemness. Recent studies have identified the APBB1/KAT5/ axis in human SSCs, where APBB1 interacts with the acetyltransferase KAT5 to suppress GDF15 expression, inhibiting differentiation and supporting self-renewal decisions essential for male fertility. Feedback loops involving RNA-binding proteins further favor renewal by suppressing pro-differentiation pathways. Nanos2, downstream of GDNF signaling, binds specific mRNAs to promote their degradation, thereby repressing meiotic and somatic programs to maintain the undifferentiated spermatogonial state. Lin28, expressed in undifferentiated spermatogonia, enhances self-renewal by regulating let-7 microRNAs and promoting proliferation through pathways like PI3K/Akt, marking progenitor populations that contribute to . These interconnected mechanisms collectively ensure persistence, with niche-derived signals briefly influencing transitions to states as needed.00352-7)

Differentiation Processes

Differentiation of spermatogonial stem cells (SSCs) is initiated by (), which promotes the transition from an undifferentiated state to committed progenitors by inhibiting NANOS2, a of the stimulated by 8 (STRA8). This -mediated downregulation allows STRA8 expression, a critical regulator that coordinates the timing of spermatogonial differentiation and subsequent meiotic entry. In humans, this process synchronizes with the approximately 64-day spermatogenic cycle, ensuring continuous production. In , the differentiation proceeds through distinct stages, beginning with undifferentiated type A spermatogonia (including single A_s, paired A_pr, and aligned A_al cells) that commit to differentiating spermatogonia upon RA exposure. These undergo sequential mitotic divisions to form A2, A3, A4, , and type B spermatogonia, culminating in preleptotene spermatocytes that enter . In humans, the process is analogous but uses a different morphological , such as A_pale to and type B spermatogonia. Accompanying these morphological changes are dynamic alterations in , including a progressive remodeling of the methylome with global hypomethylation in primary spermatocytes to facilitate required for meiotic progression. These epigenetic shifts, particularly at imprinting control regions and transposon elements, ensure proper silencing of pluripotency genes and activation of differentiation-specific pathways. Key regulatory switches involve from Sertoli cells, where (SCF) binds to the receptor on differentiating spermatogonia, driving their proliferation and survival beyond the stem cell pool. This SCF/ interaction is upregulated by RA, amplifying mitotic divisions in a stage-specific manner. In mice, RA pulses occur every 8.6 days, propagating as waves along seminiferous tubules to temporally coordinate these transitions and prevent desynchronization. Barriers to efficient arise from excessive self-renewal signals, such as overexpression of glial cell line-derived neurotrophic factor (GDNF) from Sertoli s, which accumulates undifferentiated spermatogonia into clusters that fail to progress, leading to and in mouse models. This imbalance disrupts the delicate equilibrium between maintenance and commitment, highlighting the need for precise spatiotemporal control of niche factors.

Experimental Techniques

Isolation and Culture Methods

Isolation of spermatogonial stem cells (SSCs) from testicular typically involves enzymatic to yield a viable single-cell suspension. A two-step process using collagenase for initial fragmentation followed by or dispase for further cell dispersion is widely adopted, effectively enriching for undifferentiated cells while minimizing contamination. Purification of the SSC fraction relies on immunomagnetic or fluorescence-based sorting techniques targeting specific surface markers. (MACS) with anti-THY1 or anti-GFRα1 antibodies achieves high-purity isolation of SSCs in , with GFRα1 selection yielding significant enrichment (up to ~25%) of SSC-containing populations in models. Fluorescence-activated cell sorting (FACS) enables multi-marker selection, such as combinations of THY1, ITGA6, and EPCAM, to further refine SSC populations across species. In vitro culture of isolated SSCs emphasizes maintenance of self-renewal through defined, serum-free media formulations. Essential supplements include glial cell line-derived neurotrophic factor (GDNF) at 40 ng/mL and at 10 ng/mL, often combined with GFRα1 ligands to promote proliferation; these conditions support rodent SSC expansion for over six months on mitotically inactivated feeder layers like STO fibroblasts. Recent optimizations incorporate glycolysis-promoting metabolites in feeder-free setups, enhancing SSC yield and viability without loss of germline potential. Advances in SSC culture have shifted toward three-dimensional () systems to replicate the testicular niche more accurately. Alginate-based s and decellularized scaffolds facilitate SSC attachment, survival (up to 74% viability), and differentiation, outperforming traditional 2D monolayers by preserving cell-cell interactions and reducing . These approaches, including co-cultures with Sertoli cells, have enabled short- to medium-term propagation of SSCs from fetal or prepubertal , though full long-term remains elusive. As of 2025, novel microneedle-based systems have enabled whole testicular cultivation, supporting spermatogonial proliferation and improving models. SSC stemness in culture is functionally validated through transplantation assays, where cells are microinjected into recipient testes to confirm colonization and spermatogenic recovery. In rodent systems, successful cultures demonstrate over 100 population doublings while retaining stemness markers such as GFRA1 and exhibiting low tumorigenic potential upon transplantation, as validated by purification and culture methods. Key limitations in SSC isolation and culture arise from interspecies variations in marker expression and niche dependencies. Human SSCs prove particularly recalcitrant to long-term propagation due to the complexity of their somatic microenvironment, often resulting in differentiation or quiescence rather than sustained self-renewal observed in mice.

Transplantation Procedures

Transplantation of spermatogonial stem cells (SSCs) involves the surgical delivery of isolated donor cells into the seminiferous tubules of a recipient testis to restore spermatogenesis. The procedure was first demonstrated in mice through microinjection of donor germ cells directly into the seminiferous tubules, achieving colonization and production of donor-derived sperm capable of germline transmission. In larger animals, including primates and humans, ultrasound-guided injection into the rete testis or efferent ducts has become the preferred technique, allowing for more precise and less invasive delivery of cell suspensions while minimizing damage to testicular architecture. This method, pioneered in non-human primates, enables the infusion of larger volumes (up to several milliliters) and has been adapted for clinical settings. Recipient preparation is critical to create space for donor cell engraftment by depleting endogenous SSCs. , a chemotherapeutic alkylating agent, is commonly administered systemically to induce sterility in recipient animals, such as mice or non-human primates, by selectively targeting while sparing Sertoli cells and the testicular architecture. Optimal dosing, typically 25-40 mg/kg in , achieves near-complete germ cell ablation within 4-6 weeks, enhancing donor colonization rates. In human applications, autologous transplantation avoids the need for full depletion, focusing instead on patients with preserved but non-functional germ cell niches, such as prepubertal boys with cancer who have cryopreserved testicular tissue. An ongoing phase I (NCT04452305) evaluates ultrasound-guided injection of autologous SSCs in such patients to assess safety and feasibility. Post-transplantation outcomes include donor colonization of the tubules, restoration of , and production of functional . In mice, colonization efficiency typically ranges from 5% to 12%, with donor-derived spermatozoa appearing within 2-3 months and successfully transmitting donor to . In busulfan-treated rhesus monkeys, SSC transplantation has regenerated complete , yielding fertilization-competent that supported development . These results underscore the procedure's potential to restore across species, though efficiency varies with donor quality, recipient age, and injection precision. Key risks associated with SSC transplantation include immune rejection in allogeneic settings and potential transmission of malignancies from donor cells. Allogeneic transplants between genetically mismatched donors and recipients can provoke immune responses against donor SSCs, leading to poor engraftment unless is used, which carries additional toxicities. For donors with a , such as , there is a of transferring contaminating tumor cells, necessitating rigorous purging protocols like multicolor to enrich for pure SSCs and exclude malignant populations. Autologous approaches mitigate these risks but require viable cryopreserved cells from the recipient's own tissue.

Cryopreservation Strategies

Cryopreservation of spermatogonial stem cells (SSCs) is essential for preserving male fertility, particularly in cases of gonadotoxic treatments, and involves strategies to minimize cellular damage from ice crystal formation and osmotic stress during freezing and thawing. Two primary approaches are employed: slow-freezing for dispersed SSC suspensions and vitrification for intact testicular biopsies. Slow-freezing uses controlled cooling rates with permeable cryoprotectants like dimethyl sulfoxide (DMSO) to allow gradual dehydration and prevent intracellular ice, while vitrification relies on ultra-rapid cooling with high cryoprotectant concentrations to achieve a glass-like state, avoiding ice damage in tissue fragments. In slow-freezing protocols for dispersed , typically from models, cells are suspended in containing 1.4 /L DMSO, equilibrated at , and cooled at an optimal rate of -1°C/min to -80°C before storage in . This method yields post-thaw viability of 65-87% in SSCs, as assessed immediately after thawing. For human applications, which have been explored since the early 2000s for pediatric patients, similar DMSO-based slow-freezing of testicular tissue achieves around 74% viability, supporting SSC post-thaw. , suited for whole testicular biopsies (0.3-1.5 mm³), involves exposure to mixtures like 1.1 /L DMSO, 1.3 /L ethylene glycol, and 250 mmol/L , followed by direct plunging into , preserving human SSC functionality for transplantation. Viability is commonly evaluated using trypan blue exclusion staining, which distinguishes live from membrane-compromised cells, while functional recovery is confirmed through transplantation assays measuring SSC colonization efficiency. In models, cryopreserved SSCs demonstrate 70-85% retention of colonization potential in recipient testes, enabling restoration. These assessments highlight that while viability is high, long-term niche interactions post-thaw remain challenging to replicate, potentially affecting cues. Recent advances include the incorporation of as a non-permeable additive to enhance cryoprotection without . Studies from 2024 report that 100-200 mmol/L supplementation in freezing media improves and bovine SSC recovery by reducing and , boosting post-thaw by up to 49% in models. Despite these improvements, challenges persist in maintaining SSC-niche-like conditions during thawing to ensure full functional integration upon transplantation.

Applications and Advances

Therapeutic Uses in Infertility

Spermatogonial stem cell (SSC) transplantation represents a primary therapeutic application for restoring in prepubertal boys who face infertility risks from gonadotoxic , such as for or other cancers. In this autologous approach, testicular tissue containing SSCs is cryopreserved prior to treatment and later transplanted back into the patient's testes to repopulate the and enable post-puberty. This method addresses the inability to bank mature in prepubertal patients, offering a potential lifeline for survivors who otherwise risk permanent . SSC-based therapies also hold promise for treating non-obstructive in genetic disorders like (47,), where focal areas of may exist despite overall testicular failure. Autologous transplantation of enriched SSCs from these regions could regenerate production, bypassing the limitations of microdissection testicular extraction, which succeeds in only 40-50% of cases. Preclinical studies have demonstrated successful propagation and transplantation of XXY SSCs , supporting their potential to colonize recipient tubules and restore . Clinical progress includes ongoing Phase I trials, such as the University of Pittsburgh's study initiated in November 2023, which evaluates the safety and feasibility of SSC transplantation in cancer survivors. However, ethical concerns arise with potential editing to enhance SSC viability, including risks of heritable off-target mutations and equitable access, prompting calls for stringent regulatory oversight. Adjunctive strategies, like co-culturing SSCs with bone marrow-derived mesenchymal stem cells, have improved survival in animal models through paracrine support. While proof-of-concept has been established in animal models, with donor-derived achieving up to 80% tubule colonization in mice and nonhuman primates, outcomes remain limited, with engraftment success rates below 50% due to challenges in reconstituting the testicular niche and ensuring long-term SSC maintenance. These limitations underscore the need for optimized protocols to improve colonization and minimize immune rejection in autologous settings.

Conservation and Emerging Research

Spermatogonial stem cell () banking through of gonadal tissues has emerged as a vital strategy for preserving in endangered species, enabling long-term storage of for future restoration efforts. This approach complements broader biobanking initiatives, such as those promoted by the IUCN Species Survival Commission, which emphasize the collection and cryogenic preservation of reproductive cells to support programs. For instance, advancements in SSC cryopreservation protocols have been adapted across various taxa, including mammals and birds, to safeguard populations facing extinction risks from habitat loss and . Xenotransplantation of SSCs offers a promising avenue for interspecies transmission, allowing donor cells from to be transplanted into surrogate hosts of closely related taxa to facilitate and offspring production. Successful xenogeneic transplantation has been demonstrated in models, where cells and SSCs from rare bird species colonized recipient gonads, leading to the production of donor-derived gametes. In mammals, similar techniques hold potential for species like felids and canids, though challenges in immune and colonization efficiency persist. These methods could restore in depleted populations, as explored in frameworks for preservation. Bibliometric analyses conducted in 2025 reveal a surge in SSC research, with a consistent annual publication rate of over 100 articles in recent years highlighting a shift toward advanced genomic tools, particularly single-cell sequencing (scRNA-seq) to map the human and identify progenitor states. These trends underscore a growing emphasis on elucidating the molecular signatures of SSC self-renewal and differentiation, with scRNA-seq enabling the resolution of heterogeneous subpopulations within the undifferentiated spermatogonial compartment. Novel markers such as (FGFR3) have been identified as enriched in SSC subsets via scRNA-seq, supporting further studies on purification from human testicular biopsies. Future directions in SSC research include CRISPR-Cas9-mediated gene editing to generate models of genetic diseases affecting spermatogenesis, such as corrections in mouse SSCs for heritable mutations like those in the Kit gene, demonstrating efficient homology-directed repair and transmission to offspring. Integration of SSCs with testicular organoids represents another frontier, where three-dimensional cultures recapitulate the niche microenvironment to support in vitro spermatogenesis, progressing from spermatogonial proliferation to haploid gamete formation in rodent and human-derived systems. These organoid platforms, often incorporating somatic support cells, provide scalable models for studying germline development without animal models. Despite these advances, applications in face significant challenges, including species-specific adaptations in niche signaling and tolerances that hinder protocol translation across taxa. For example, variations in composition and hormonal requirements complicate success rates in non-rodent mammals. Ethical concerns surrounding wildlife germline modification are also prominent, encompassing risks of unintended ecological impacts, during transplantation procedures, and the moral implications of altering genetic lineages in endangered populations without comprehensive long-term safety data. Addressing these requires interdisciplinary guidelines to balance innovation with responsible stewardship.

References

  1. [1]
    Spermatogonial stem cells: Current biotechnological advances in ...
    Spermatogonial stem cells (SSCs) are the germ stem cells of the seminiferous epithelium in the testis. Through the process of spermatogenesis, they produce ...
  2. [2]
    Defining Characteristics of Mammalian Spermatogenic Stem Cells
    Cells destined to become the male germline stem cell population, also referred to as spermatogonial stem cells (SSCs), originate in the embryo from embryonal ...
  3. [3]
    The nature and dynamics of spermatogonial stem cells | Development
    Sep 1, 2017 · Spermatogonial stem cells (SSCs) are crucial for maintaining spermatogenesis throughout life, and understanding how these cells function has important ...
  4. [4]
    Spermatogenic Stem Cells: Core Biology, Defining Features, and ...
    Oct 11, 2024 · The actions of spermatogenic stem cells (SSCs) provide the foundation for continual spermatogenesis and regeneration of the cognate lineage.
  5. [5]
    Spermatogonial stem cells - PMC - PubMed Central - NIH
    Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis.
  6. [6]
    Spermatogonial stem cell regulation and spermatogenesis - Journals
    May 27, 2010 · This article will provide an updated review of spermatogonial stem cells and their role in maintaining the spermatogenic lineage.Spermatogenic lineage... · Spermatogonial stem cell... · Dissecting the molecular...
  7. [7]
    Review Spermatogenesis in humans and its affecting factors
    The estimated daily sperm production per man ranges from 150 to 275 million spermatozoa [3], [5].
  8. [8]
    Roles of Spermatogonial Stem Cells in Spermatogenesis and ...
    May 12, 2022 · The SSCs are the resident stem cell population that resides at the basal membrane of seminiferous tubules of the testis (4, 5).
  9. [9]
    THE GERMLINE STEM CELL NICHE UNIT IN MAMMALIAN TESTES
    In addition, the position of SSCs in the basal compartment of the seminiferous epithelium, below Sertoli cell tight junctions that form the blood-testis- ...
  10. [10]
    The central role of Sertoli cells in spermatogenesis - PubMed
    Sertoli cells facilitate the progression of germ cells to spermatozoa via direct contact and by controlling the environment milieu within the seminiferous ...
  11. [11]
    Histology, Leydig Cells - StatPearls - NCBI Bookshelf - NIH
    Leydig cells are essential testicular cells, located between seminiferous tubules, and are the primary source of testosterone in males.Introduction · Structure · Function · Histochemistry and...
  12. [12]
    Peritubular myoid cells in the testis: their structure and function
    Myoid cells have been shown to be contractile, involved in the transport of spermatozoa and testicular fluid in the tubule. Several substances (prostaglandins, ...
  13. [13]
    Male germ-line stem cell potential is predicted by morphology of ...
    Spermatogenesis is initiated in the testes of postnatal mammals when quiescent gonocytes resume proliferation, migrate to the seminiferous tubule basement ...
  14. [14]
    Temporal-Spatial Establishment of Initial Niche for the Primary ...
    During neonatal testis development, centrally located gonocytes migrate to basement membrane of the seminiferous cords, where physical contact with a niche ...
  15. [15]
    Spermatogonium - an overview | ScienceDirect Topics
    Spermatogonia were first identified as separate entities from the Sertoli cells by von Ebner in 1871 (159), but the term spermatogonia was first applied to this ...Missing: nomenclature | Show results with:nomenclature
  16. [16]
    The spermatogonial stem cell population in adult rats. I ... - PubMed
    The spermatogonial stem cell population in adult rats. I. Their morphology, proliferation and maturation. Anat Rec. 1971 Mar;169(3):533-57. doi: ...Missing: Oakberg | Show results with:Oakberg
  17. [17]
    Spermatogonial stem cells and spermatogenesis in mice, monkeys ...
    Apr 21, 2018 · These rare cells are called the Asingle spermatogonia (As), which divide once every three days and make up about 0.03% of the total germ cells ...Missing: percentage | Show results with:percentage
  18. [18]
    Identification of Spermatogonial Stem Cell Subsets by ...
    In primates, including humans, spermatogonial stem cells are among Adark (Ad) and Apale (Ap) spermatogonia [1]. Ad and Ap spermatogonia were identified on ...
  19. [19]
    Microenvironment of spermatogonial stem cells: a key factor in the ...
    Sep 11, 2024 · Spermatogonial stem cells (SSCs) play a crucial role in the male reproductive system, responsible for maintaining continuous spermatogenesis ...
  20. [20]
    Spermatogonial stem cell technologies: applications from human ...
    While functional evidence is sparse, both Adark, and Apale are thought to possess stem cell activity, with Adark spermatogonia being the reserve, non- or slow- ...
  21. [21]
    Spermatogonial stem cells share some, but not all, phenotypic and ...
    Spermatogonial stem cells (SSCs) are responsible for maintaining spermatogenesis throughout life in the male by continuous production of daughter cells.Missing: review | Show results with:review
  22. [22]
    Functional assessment of spermatogonial stem cell purity in ...
    Apr 3, 2018 · Historically, research in spermatogonial biology has been hindered by a lack of validated approaches to identify and isolate pure populations of ...Missing: challenges universal
  23. [23]
    Regulation of the Spermatogonial Stem Cell Niche - PMC
    Spermatogonial stem cells reside in the basal part of the seminiferous epithelium. These cells are the only stem cells in the body that undergo self-renewal ...<|control11|><|separator|>
  24. [24]
    Regulation of GDNF expression in Sertoli cells - PubMed Central - NIH
    Sertoli cells regulate male germ cell proliferation and differentiation and are a critical component of the spermatogonial stem cell (SSC) niche, ...
  25. [25]
    A laminin-based local regulatory network in the testis that supports ...
    In adult rat testes, the basement membrane is structurally constituted by laminin and collagen chains that lay adjacent to the blood-testis barrier (BTB).
  26. [26]
    What do we know about blood-testis barrier? current understanding ...
    Spermatogonial stem cells alone are not sufficient to re-initiate spermatogenesis in the rat testis following adjudin-induced infertility. Int. J. Androl ...
  27. [27]
    Effects of Aging and Niche Microenvironment on Spermatogonial ...
    We test the hypothesis that aging results in uncorrected defects in stem cell and/or niche function, which lead to system failure.
  28. [28]
    Follicle-stimulating hormone signaling in Sertoli cells: a licence to ...
    Jul 2, 2022 · Follicle-stimulating hormone signaling is essential for the initiation and early stages of spermatogenesis.
  29. [29]
    Decoding the spermatogonial stem cell niche under physiological ...
    Aug 4, 2023 · Decoding the spermatogonial stem cell niche under physiological and recovery conditions in adult mice and humans
  30. [30]
    Growth factors essential for self-renewal and expansion of mouse ...
    Nov 16, 2004 · The spermatogenic system depends on stem cells, which have the ability to self-renew and generate a large number of differentiated germ cells in ...
  31. [31]
    RET receptor signaling: Function in development, metabolic disease ...
    Signaling via the GDNF/GFRα1/RET ternary complex plays crucial roles in the ... self-renewal of spermatogonial stem cells. In addition, another ligand ...
  32. [32]
    Spermatogonial Stem Cell Numbers Are Reduced by ... - PubMed
    Mar 9, 2021 · Spermatogonial Stem Cell Numbers Are Reduced by Transient Inhibition of GDNF Signaling but Restored by Self-Renewing Replication when Signaling ...
  33. [33]
    Essential Regulation of Spermatogonial Stem Cell Fate Decisions ...
    Mar 27, 2025 · Our findings highlight an essential role of APBB1/KAT5/GDF15 in governing human SSC fate decisions and maintaining normal spermatogenesis.
  34. [34]
    The pluripotency factor LIN28 marks undifferentiated spermatogonia ...
    Jun 29, 2009 · In a normal situation, the actual stem cells undergo self-renewal and give rise to transit cells that further divide to become terminally ...
  35. [35]
    The Role of Retinoic Acid in Spermatogenesis and Its Application in ...
    NANOS2 can silence Stra8, a gene involved in spermatogonial differentiation and meiosis, and RA can promote spermatogonial differentiation by inhibiting NANOS2 ...
  36. [36]
    Stimulated by retinoic acid gene 8 (Stra8) plays important roles ... - NIH
    Stimulated by retinoic acid gene 8 (Stra8) is a key molecule involved in meiosis initiation and plays an important role in spermatogenesis.
  37. [37]
    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 ...
  38. [38]
    Recent Update on Retinoic Acid-Driven Initiation of Spermatogonial ...
    These six mitotic divisions occur over a span of exactly 8.6 days in mice testes (Griswold, 2016). ... Cycles, Waves, and Pulses: Retinoic Acid and the ...
  39. [39]
    Mechanisms regulating mammalian spermatogenesis and fertility ...
    Spermatogonial differentiation in rhesus monkeys produces type B1 spermatogonia that undergo further rounds of division to produce B2, B3, and B4 cells ...
  40. [40]
    Genome-wide DNA methylation changes in human spermatogenesis
    May 16, 2024 · We found that spermatogenesis is associated with remodeling of the methylome, comprising a global decline in DNA methylation in primary spermatocytes followed ...
  41. [41]
    DNA methylation and gene expression dynamics during ...
    Aug 20, 2015 · To understand the regulation of spermatogonial stem cell formation and differentiation, we examined the DNA methylation and gene expression ...
  42. [42]
    c-kit and its related genes in spermatogonial differentiation - PMC
    Kitl, also called SCF, is produced in the Sertoli cells and is a cytokine essential for haematopoiesis, melanogenesis and development of germ cells. Kitl has ...
  43. [43]
    Retinoic acid regulates Kit translation during spermatogonial ...
    Jan 1, 2015 · Retinoic acid induces Sertoli cell paracrine signals for spermatogonia differentiation but cell autonomously drives spermatocyte meiosis.
  44. [44]
    Cycles, waves, and pulses: Retinoic acid and the organization of ...
    ... 8.6 days in the mouse. The spermatogenic wave initially described in detail by Perey et al was defined as a series of adjacent tubule segments that included ...
  45. [45]
    The In Vivo Response of Stem and Other Undifferentiated ... - NIH
    Over expression of GDNF resulted in the formation of clusters of undifferentiated spermatogonia many of which died by apoptosis, resulting in an infertile adult ...Missing: overactive | Show results with:overactive<|control11|><|separator|>
  46. [46]
    Regulation of Cell Fate Decision of Undifferentiated Spermatogonia ...
    Aug 10, 2025 · Gene-targeted mice with one GDNF-null allele show depletion of stem cell reserves, whereas mice overexpressing GDNF show accumulation of ...
  47. [47]
    Recent advances in isolation, identification, and culture of ... - NIH
    Recent advances include research on the biology, isolation, and culture of SSCs, and the use of two-step enzymatic digestion for isolation.
  48. [48]
    Identification of a Technique Optimized for the Isolation of ...
    Overall, our results indicate that MACSGFRα1 is an appropriate purification technique for the isolation of SSCs from mouse testicular cells. Keywords: ...
  49. [49]
    THY1 is a conserved marker of undifferentiated spermatogonia in ...
    Following enzymatic digestion and MACS isolation, THY1+ and unselected total testis cell populations were resuspended in PBS and labeled with PKH26 red ...<|separator|>
  50. [50]
    FACS and MACS sorting strategies to isolate and enrich human ...
    THY1, ITGA6 and EPCAM are cell surface markers that have each been used to isolate and enrich spermatogonial stem cells in other species (15, 16, 25, 75).
  51. [51]
    Spermatogonial stem cells† | Biology of Reproduction
    Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis.
  52. [52]
    GDNF-expressing STO feeder layer supports the long-term ... - Nature
    Nov 9, 2016 · A GDNF-expressing STO feeder could support undifferentiated mouse spermatogonia propagation in vitro for three months without the refreshment of recombinant ...
  53. [53]
    Glycolysis-Optimized Conditions Enhance Maintenance of ...
    May 9, 2017 · Improved serum- and feeder-free culture of mouse germline stem cells ... GDNF-dependent and FGF2-dependent mouse spermatogonial stem cell ...
  54. [54]
    Recent advances in developing 3D culture systems of ... - NIH
    Oct 30, 2023 · Modern techniques developed advanced 3-dimensional co-cell cultures, and differentiation of SSC and their precursor cells have, in any case, ...
  55. [55]
    Culture and transplantation of spermatogonial stem cells
    Spermatogonial stem cells (SSCs) provide the foundation for spermatogenesis. In mice, SSCs comprise 0.02–0.03% of total germ cells in testes and are thought to ...Missing: percentage | Show results with:percentage
  56. [56]
    Postnatal development of mouse spermatogonial stem cells ... - Nature
    Jan 27, 2024 · Such SSC cultures may also effectively lead to a demise of tumorigenic cells prior to transplantation through the environment preferable for SSC ...
  57. [57]
    Recent advances of in vitro culture systems for spermatogonial stem ...
    This review summarizes the recent progress in the development of the culture system and possible challenges in establishing a SSC line in human and livestock ...
  58. [58]
    Prospects and problems for germ cell transplantation in the male
    Dec 24, 2001 · Therefore an alternative technique was developed for germ cell transfer into the primate testis, using ultrasound‐guided injections into the ...
  59. [59]
    Efficient and Safe Recipient Preparation for Transplantation of ...
    Recipientpreparation is of prime importance for the successful transplantation of spermatogonial stem cells (SSCs). Busulfan destroys endogenous germs cells and ...Busulfan Treatment · Testicular Cell Preparation... · Results
  60. [60]
    Optimal dose of busulfan for depleting testicular germ cells of ... - NIH
    The results suggest that a dosage of 30 mg kg −1 is optimal for the ablative treatment with busulfan used to prepare the recipient mice.
  61. [61]
    Spermatogonial Stem Cell (SSC) Transplant and Testicular Tissue ...
    Spermatogonial stem cell transplantation through ultrasound guided rete testis injection and testicular tissue grafting will be performed for participants ...
  62. [62]
    Propagation of Human Spermatogonial Stem Cells In Vitro
    Nov 18, 2009 · From studies of transplantation of autologous animal spermatogonial stem cells, we know that the efficiency of colonization is between 5% and 12 ...
  63. [63]
    Spermatogonial Stem Cell Transplantation in Large Animals - MDPI
    Mar 24, 2021 · ... efficacy of SSC homing. The efficiency of SSC colonization is 4–10 times higher in immature testis than in mature testis and also ...
  64. [64]
    Eliminating malignant contamination from therapeutic human ... - JCI
    Mar 15, 2013 · Spermatogonial stem cell (SSC) transplantation has been shown to restore fertility in several species and may have application for treating ...
  65. [65]
  66. [66]
    Cryopreservation and culture strategies for testicular tissue and cells ...
    The two most widely used cryopreservation methods are slow freezing and vitrification. Few reports used a modified technique called rapid freezing (Table 1) (5, ...
  67. [67]
    Cryopreservation and culture strategies for testicular tissue and cells ...
    Jul 16, 2025 · Therefore, this review aims to summarize and critically assess current cryopreservation and culture strategies for testicular tissues and cells ...
  68. [68]
    Cryopreservation of spermatogonial stem cells - PubMed
    Apr 22, 2025 · This review summarizes the effects of multiple cryoprotectants in SSC cryopreservation, providing ideas for optimizing SSC freezing techniques ...
  69. [69]
    Advancements in fertility preservation strategies for pediatric male ...
    Apr 18, 2024 · Wyns et al. discovered that slow freezing can preserve the proliferation capacity of spermatogonial cells even after 6 months of xenografting, ...
  70. [70]
    Reestablishment of spermatogenesis after more than 20 years of ...
    May 10, 2022 · These findings strongly suggest that prolonged cryopreservation can impact the success of transplantation to produce spermatogenesis, which may ...
  71. [71]
    A review on the functional roles of trehalose during cryopreservation ...
    The inclusion of trehalose in cryopreservation extender has been widely reported to enhance antioxidant activity, decrees the oxidative stress and improve the ...
  72. [72]
    Fertility Preservation and Restoration Options for Pre-Pubertal Male ...
    Jun 16, 2022 · Furthermore, it has been estimated that only 10% of transplanted spermatogonial stem cells are able to form colonies (149). Such cells are rare, ...
  73. [73]
    In vitro propagation of XXY human Klinefelter spermatogonial stem ...
    At the time of transplantation, only SSCs are expected to migrate to the basal membrane of the seminiferous tubules and restore spermatogenesis in azoospermic ...
  74. [74]
    The Evidence for Fertility Preservation in Pediatric Klinefelter ...
    Aug 2, 2021 · The concept of in vitro spermatogonial stem cell (SCC) propagation and auto-transplantation has been explored recently to provide another ...
  75. [75]
    Risks and benefits of human germline genome editing - NIH
    The first possible application is infertility treatment. Oocyte editing and spermatogonial editing could be an option for patients suffering from genetic ...
  76. [76]
    Mesenchymal stem cells promote spermatogonial stem/progenitor ...
    Jul 7, 2022 · We hypothesized that an ALI co-culture system based on syngeneic BM-MSC could support survival, expansion and differentiation of the SSPC pool ...
  77. [77]
    [EPUB] Cellular functions of spermatogonial stem cells in relation to JAK ...
    Feb 13, 2024 · After GDNF withdrawal, there is increased activation of STAT3 and Neurog3 genes in spermatogonial cell cultures, with suppression upon re- ...Missing: boost | Show results with:boost
  78. [78]
    Autologous spermatogonial stem cell transplantation in man
    Jan 10, 2008 · The spermatogonial stem cells are the male germline stem cells. They can self renew to maintain the stem cell population and produce large ...
  79. [79]
    Spermatogonial Stem Cell-Based Therapies: Taking Preclinical ...
    Apr 3, 2022 · Currently, there is a survival rate of 80% among pediatric cancer patients in Europe (1), leading to a large population of childhood cancer ...Introduction · Cryopreservation · SSC-Based Techniques · Safety
  80. [80]
    State of the art and application of gamete and reproductive tissue ...
    In this review, we examine the status of cryopreservation protocols for gametes, embryos, and gonadal tissues across various wildlife taxa, highlighting recent ...
  81. [81]
    [PDF] 2024-2025 Report of the IUCN Species Survival Commission and ...
    The IUCN Species Survival Commission. (SSC) is a science-based network of thou- sands of volunteer experts from almost every country of the world, all working.
  82. [82]
    Spermatogonial stem cell technologies: applications from human ...
    Alternatively, donor SSCs could be transplanted into a recipient from a closely related species (xenotransplantation) to facilitate in vivo spermatogenesis and ...
  83. [83]
    Conservation of Avian Germplasm by Xenogeneic Transplantation ...
    In this study, we aimed to exploit the potential of germ cell xenotransplantation as an alternative tool for preserving germplasm of endangered birds. The study ...
  84. [84]
    Advances in spermatogonial stem cell research: A systematic review ...
    Aug 8, 2025 · In this study, we used bibliometric analysis to assess worldwide research trends in the SSCs area based on publication outputs, co-authorships ...
  85. [85]
    A single-cell view of spermatogonial stem cells - PMC - NIH
    Sep 17, 2020 · Here, we discuss what scRNAseq analysis has revealed about SSC-precursor cells, the initial generation of SSCs during perinatal development, and ...Missing: bibliometric 2025 trends
  86. [86]
    Human spermatogonial stem cells scrutinized under the single ... - NIH
    Feb 7, 2019 · ... FGFR3). Novel SSC marker genes were also identified, many of which were verified as candidate SSC markers by in situ RNA hybridization and ...
  87. [87]
    Correction of a genetic disease by CRISPR-Cas9-mediated gene ...
    Dec 5, 2014 · Our results demonstrate efficient gene editing in mouse SSCs by the CRISPR-Cas9 system, and provide the proof of principle of curing a genetic disease via gene ...Results · Crispr-Cas9-Mediated Genetic... · Electroporation Of Sscs
  88. [88]
    Genome editing in mouse spermatogonial stem cell lines targeting ...
    In spermatogonial stem cells (SSCs), CRISPR/Cas9 has proven to be an effective tool for elucidating the genetic mechanisms underlying spermatogenesis and ...
  89. [89]
    Spermatogonial Stem Cells and In Vitro Spermatogenesis - NIH
    There are two general IVS approaches: organotypic cultures and “bottom-up” organoids. Organotypic cultures consist of testicular tissue cultured in vitro for ...
  90. [90]
    Genetic engineering of animals: Ethical issues, including welfare ...
    The genetic engineering of animals has increased significantly in recent years, and the use of this technology brings with it ethical issues, some of which ...Missing: adaptations germline SSCs
  91. [91]
    Employing induced Pluripotent Stem Cells for Primordial Germ Cells ...
    Another key ethical concern is the welfare of the animals involved. Biotechnological techniques could lead to unforeseen health issues or suffering in ...