Fact-checked by Grok 2 weeks ago

Somatic cell

A somatic cell is any cell in a that is not a reproductive cell, such as a or , and instead forms the structural and functional building blocks of the body's tissues and organs. In humans, somatic cells are diploid, containing two complete sets of 46 chromosomes—one inherited from each parent—arranged in 23 pairs. These cells arise from the through repeated mitotic divisions and differentiate into specialized types, such as muscle, , or cells, to support organismal function and maintenance. Somatic cells divide via , a process that produces two genetically identical daughter cells to enable growth, repair, and replacement of tissues throughout an organism's life. Unlike germ cells, which undergo to produce haploid gametes for , somatic cells maintain their diploid state and do not contribute directly to . Mutations occurring in somatic cells, known as , can lead to conditions like cancer but are not passed on to offspring since they do not affect the . Among cells, a subset known as somatic stem cells plays a crucial role in regeneration and tissue by self-renewing and differentiating into multiple cell types as needed. These cells are found in various adult tissues, such as and , and their regulated activity is essential for preventing diseases associated with uncontrolled proliferation, including tumors. Research into somatic cells has advanced fields like , including techniques such as used in . Notable advancements include the development of induced pluripotent stem cells (iPSCs) from somatic cells, enabling patient-specific regenerative therapies.

Definition and Characteristics

Definition

Somatic cells are any cells in multicellular organisms that constitute the , excluding the reproductive germ cells such as and eggs. These cells form the structural and functional components of tissues and organs, performing essential roles in , , and response to environmental stimuli. The term "" originates from the Greek word sōma, meaning "body," reflecting their role in composing the organism's physical structure. The concept of somatic cells was formalized in the late by biologist , who introduced the distinction between somatic cells and germ cells to explain . In his 1892 essay Das Keimplasma, Weismann proposed that only germ cells carry hereditary information across generations, while somatic cells do not contribute to inheritance, thereby challenging theories of acquired trait transmission. This framework laid the groundwork for modern by emphasizing the separation of somatic and lineages. In , somatic cells are typically diploid, containing two sets of chromosomes (2n)—one set from each parent—which supports their stability and specialized functions through ; however, somatic cells in and some other organisms can be polyploid. The and early undifferentiated embryonic cells exhibit totipotency, the ability to differentiate into any , including extra-embryonic tissues; as cells differentiate into the somatic lineage, this potential diminishes, leading to multipotent stem cells in tissues that can form multiple related cell types, or unipotent cells in mature states that produce only one type. Representative examples of somatic cells include neurons, which transmit electrical signals in the ; muscle cells, responsible for and ; and epithelial cells, which line surfaces and cavities for protection and absorption.

Distinction from Germ Cells

Somatic cells and germ cells represent two distinct s in multicellular organisms, differing primarily in their roles and fates. Somatic cells constitute the majority of the body's non-reproductive tissues, functioning to construct, maintain, and repair structures such as muscles, organs, and throughout an individual's life. In contrast, germ cells—encompassing primordial cells that develop into and oocytes—are specialized for , serving as the vehicles for transmitting genetic material across generations. This functional dichotomy ensures that somatic cells support immediate survival and , while germ cells preserve continuity. A pivotal distinction lies in , governed by the , which prevents the transmission of somatic modifications to offspring. Proposed in August Weismann's , this barrier maintains that the is isolated from somatic influences, such that acquired changes in body cells, like environmental damage or aging effects, do not alter the genetic information passed to progeny. Only variations within germ cells can contribute to inheritance, safeguarding evolutionary stability by excluding non-adaptive somatic alterations from the . Both cell types originate from the diploid following fertilization, initially sharing a common developmental pathway through mitotic divisions that generate the early . However, somatic cells persist in diploid form and proliferate exclusively via to differentiate into the organism's various tissues. Germ cells, set aside early in embryogenesis as primordial germ cells, eventually undergo to reduce their and produce haploid gametes capable of fertilization. These differences have profound implications for and : alterations in somatic cells, such as oncogenic mutations, can drive conditions like cancer within the affected individual but remain confined to that generation due to the , lacking evolutionary heritability. This non-transmissible nature underscores why somatic pathologies do not propagate genetically, focusing therapeutic efforts on individual-level interventions rather than lineage-wide effects.

Evolutionary Origins

Emergence in Multicellularity

The emergence of somatic cells coincided with the transition from unicellular to multicellular eukaryotes, occurring at least 1.6 billion years ago (as of 2024) as cells began forming aggregates with specialized functions. This evolutionary shift arose from unicellular ancestors through mechanisms like incomplete , where daughter cells remained attached, fostering cooperation and division of labor that enabled non-reproductive cells to support organismal functions such as and nutrient uptake. In volvocine green algae, such as , this process is exemplified by the evolution of sterile cells alongside reproductive gonidia, driven by genetic changes like the co-option of the regA gene to repress reproduction in somatic lineages, allowing specialization for tasks like swimming while larger cells focus on . These algae illustrate how somatic cells lost totipotency—the ability to develop into any —to form structured tissues, a key event in early multicellularity that promoted efficient and organismal integrity. Fossil evidence from the Ediacaran biota, approximately 600 million years ago, reveals embryo-like microfossils with differentiated cell types, including somatic cells that exhibit reduced developmental potential compared to totipotent precursors, indicating early tissue formation in complex multicellular forms. This specialization conferred advantages such as larger body sizes, enhanced environmental resilience, and improved survival via intercellular cooperation, stabilizing multicellular assemblies against cheaters and enabling greater ecological success. In primitive multicellular organisms, this often involved an initial distinction between somatic and germ-like cells to protect reproductive lineages.

Somatic-Germ Line Separation

The separation of somatic and germ lines represents a pivotal evolutionary innovation in multicellular organisms, ensuring that hereditary information is insulated from somatic modifications. proposed the germ plasm theory in the late 1880s, positing that an immutable within germ cells is distinct from the modifiable , thereby preventing the inheritance of acquired somatic traits. This theory, detailed in his 1893 work The Germ-Plasm: A Theory of Heredity, emphasized that only changes to the could be heritable, laying foundational groundwork for modern . Contemporary understanding supports Weismann's framework through epigenetic mechanisms, where the soma experiences reversible modifications like DNA methylation and histone alterations that are largely reset in the germline to maintain genomic integrity. Evolutionary models of germ line specification diverge into deterministic and inductive strategies. In deterministic models, germ cells are set aside early via inheritance of maternal determinants, as seen in mammals where primordial germ cells are segregated during early embryogenesis to avoid somatic influences. Conversely, inductive models involve later specification through signaling from somatic tissues, exemplified in fruit flies (Drosophila melanogaster), where germ cells form in response to environmental cues post-fertilization. Evidence for this separation draws from comparative biology across and , revealing conserved patterns that demarcate germ lines. For instance, piwi genes, encoding family proteins, exhibit germline-specific expression and function in silencing transposable elements to safeguard stability, a role conserved from flies to mammals. In , however, the separation is incomplete, with germ cells arising late from progenitors in floral meristems, allowing potential transmission of somatic variations. This distinction underscores the theory's implications: the -germ line barrier primarily prevents the accumulation and hereditary transmission of somatic mutations, promoting evolutionary fidelity, though its absence in enables adaptive .

Genetic Features

Chromosomes and Ploidy

Somatic cells in most animals and are diploid, possessing two complete sets of chromosomes designated as 2n , with each set consisting of homologous chromosomes—one inherited from each parent—that carry the same genes at corresponding loci. This diploid state arises from the fusion of haploid gametes during fertilization and is preserved in somatic lineages. Homologous pairs enable genetic redundancy and facilitate processes like through . The specific number of chromosomes in somatic cells varies across species, reflecting evolutionary adaptations; for instance, human somatic cells contain 46 s organized into 23 homologous pairs, including 22 pairs of autosomes and one pair of . Each features a , a constricted region of highly repetitive DNA that serves as the attachment site for spindle fibers during , dividing the into a short arm (p) and a long arm (q). At the ends of each are telomeres, specialized structures composed of repetitive DNA sequences and proteins that protect linear ends from fusion, degradation, or recognition as DNA damage. A provides a visual representation of an organism's complete set of chromosomes, typically derived from metaphase-arrested somatic cells and arranged in pairs by size, position, and banding patterns to reveal the 's organization. In somatic cells, chromosomal stability is maintained through , a process that ensures equitable distribution of the full diploid complement to daughter cells, thereby preserving genetic integrity across cell generations. While diploidy predominates, exceptions occur in certain organisms where somatic cells exhibit , involving more than two sets; for example, tetraploid (4n) somatic cells are common in durum wheat (Triticum turgidum), enhancing traits like grain size and stress tolerance. Additionally, in specialized animal somatic cells, —a modified lacking —generates polyploid nuclei; in salivary glands, this process yields polytene chromosomes with up to 1024 copies of the , supporting high transcriptional output for larval .

Somatic Mutations

Somatic mutations are alterations in the DNA sequence that occur in non-germline cells after conception, leading to genetic heterogeneity within an individual's tissues. These mutations arise spontaneously and accumulate over time, contributing to cellular diversity but also posing risks for disease. Unlike germline mutations, somatic mutations are not transmitted to offspring, as they affect only the affected cell and its descendants. The types of somatic mutations include point mutations, which are single nucleotide substitutions; insertions and deletions (indels), ranging from small-scale changes to larger structural variants; and chromosomal aberrations such as , where cells gain or lose whole , and copy number variations (CNVs). , for instance, often results from errors in chromosome segregation during , leading to imbalances in . These mutations can create patterns in tissues, where subpopulations of cells harbor distinct genetic profiles. Causes of somatic mutations are broadly categorized as environmental or endogenous. Environmental factors include exposure to ultraviolet (UV) radiation, which induces , and chemicals like those in cigarette smoke, containing over 70 known carcinogens that damage DNA bases. Endogenous causes encompass replication errors during and oxidative stress from intracellular free radicals, such as generating 8-oxoguanine lesions. The somatic mutation rate in humans is approximately 10^{-9} per per cell division in healthy tissues, though this varies by cell type and can increase in rapidly dividing populations. Detection of somatic mutations relies on advanced sequencing techniques, including whole-genome sequencing of bulk tissue samples to identify clonal expansions and for low-frequency variants in mosaic tissues. High-depth next-generation sequencing can detect mutations present in as few as 1-10% of cells, enabling the mapping of mutational landscapes in normal and diseased states. Clonal expansion, where mutated cells proliferate preferentially, often signals these events in tissues like or colon. The consequences of mutations include the formation of genetic mosaicism, which underlies conditions like McCune-Albright syndrome from activating mutations in genes, and drives non-heritable diseases such as cancer through activation or tumor suppressor inactivation. In cancer, for example, accumulated mutations in epithelial cells from smokers can exceed 10-fold higher rates, promoting uncontrolled growth. These changes also contribute to aging by impairing function, though they do not alter the stable diploid of unaffected cells. Overall, somatic mutations highlight the dynamic nature of the genome but underscore their role in when dysregulated.

Applications in Biotechnology

Cloning Techniques

Somatic cell nuclear transfer (SCNT) is a cloning technique in which the from a is transferred into an enucleated to create a genetically identical . The process begins with the enucleation of a mature , where its is removed using a micropipette under microscopic guidance to create a cytoplast. Next, a somatic cell , isolated from a donor animal, is inserted into the enucleated , often through or electrical to integrate the . The reconstructed is then activated chemically or electrically to initiate embryonic development, allowing it to divide and potentially form a that can be implanted into a for . The foundational experiments for SCNT were conducted by in the 1950s and early 1960s using frogs, where he demonstrated that nuclei from differentiated somatic cells, such as , could direct the development of fertile adult frogs when transplanted into enucleated eggs. Gurdon's 1962 study confirmed the viability of this approach through a survey of over 150 adult frogs derived from such nuclear transplants from single somatic cell nuclei, establishing the principle of nuclear totipotency in vertebrates. The first successful mammalian cloning via SCNT occurred in 1996 with the sheep, born from an adult cell nucleus transferred into an enucleated ovine , as reported by Wilmut and colleagues. This breakthrough extended SCNT to mammals, proving that even highly differentiated adult cells could be reprogrammed to support full-term development. SCNT has been applied in animal cloning for agricultural purposes, such as propagating elite livestock breeds to enhance traits like resistance and productivity, and in for generating transgenic models or preserving . In , cloned animals serve as superior stock to rapidly disseminate desirable without traditional breeding limitations. Therapeutically, SCNT enables the production of patient-specific embryonic stem cells for , though primarily in research settings with animals like pigs and . Despite these uses, SCNT efficiency remains low, typically 1-3% success rate from transferred embryos to live births, largely due to incomplete epigenetic where somatic chromatin fails to reset to an embryonic state, leading to developmental arrest. Recent advances, such as optimized protocols reported in 2025, aim to overcome these barriers by improving epigenetic , though live birth rates remain below 10% in most mammalian models. Key challenges in SCNT include telomere shortening, where cloned animals often inherit abbreviated from cultured somatic donors, potentially accelerating aging and contributing to health issues like premature organ failure. Imprinting errors also arise, with aberrant at imprinted loci causing placental abnormalities, large offspring syndrome, and increased abortion rates in cloned pregnancies. Genetic modifications, such as inhibitors, have been explored to mitigate these epigenetic barriers and boost fidelity.

Biobanking Practices

Biobanking of somatic cells involves the systematic collection, processing, and long-term storage of these non-reproductive cells from various tissues to support biomedical research and therapeutic applications. Somatic cells, derived from sources such as , , or tissue, are preserved to enable studies on mechanisms, , and regenerative therapies. This practice ensures the availability of high-quality, viable samples for downstream analyses, maintaining cellular integrity over extended periods. A primary for preserving cells in biobanks is , which typically employs (DMSO) as a cryoprotectant to prevent formation and cellular damage during freezing. Cells are mixed with 5-10% DMSO in a freezing medium, cooled at controlled rates (often 1°C per minute) to -80°C, and then transferred to storage at -196°C for indefinite viability. This technique is widely used for cell types including fibroblasts and cell progenitors, achieving post-thaw recovery rates of 70-90% in optimized protocols. Additionally, cells serve as starting material for generating induced pluripotent s (iPSCs) through reprogramming with factors like Oct4, , , and c-Myc; these iPSCs are then banked similarly via to create renewable resources for personalized cell models. Biobanks categorize cell collections into types such as tissue banks and cell line repositories. Tissue banks often store primary cells from blood, which contains hematopoietic stem cells (HSCs) capable of differentiating into blood lineages, or cord tissue rich in mesenchymal stem cells (MSCs) for tissue repair applications; these are processed to isolate viable cells shortly after collection. In contrast, cell line biobanks maintain immortalized lines, exemplified by cells—derived from human tissue in 1951—which proliferate indefinitely and are used as a standard for due to their robust growth and genetic stability. The practice of biobanking somatic cells originated in the post-1950s era, driven by advances in that necessitated reliable cell storage for experimental consistency. Early efforts focused on establishing tumor cell lines like to study oncogenesis, marking the shift from ad hoc sample handling to organized repositories. Modern large-scale initiatives, such as the launched in 2006, have expanded this to population-level collections, storing somatic cells from over 500,000 participants' blood samples alongside genomic and phenotypic data to facilitate epidemiological studies. Ethical considerations in somatic cell biobanking center on and protection, given the sensitive genetic information embedded in these samples. Participants must provide broad or specific for uses, often under dynamic models allowing re-contact for updates, to balance with scientific utility; however, challenges arise in ensuring comprehension of indefinite storage and potential secondary uses. risks, including re-identification from genomic data, are mitigated through protocols and secure data access controls, as mandated by regulations like the EU's GDPR. These practices underpin applications in , where banked somatic cells enable patient-specific iPSC derivation for tailored drug screening and therapies. Somatic cell biobanks also support genetic studies by providing diverse samples for variant analysis.

Genetic Engineering Methods

Genetic engineering of somatic cells involves targeted modifications to the of non-reproductive cells to treat diseases or study biological functions, distinct from editing which affects heritable traits. Early methods relied on nucleases (ZFNs), developed in the 1990s, which fuse DNA-binding domains to the to create double-strand breaks at specific sites, enabling disruption or insertion via cellular repair mechanisms. Transcription activator-like effector nucleases (TALENs), introduced around 2009-2010, improved specificity by using customizable TALE proteins from plant pathogens linked to , allowing precise editing in somatic cells with reduced off-target activity compared to ZFNs. The breakthrough came in 2012 with -Cas9, pioneered by and , which repurposes the bacterial into a programmable tool using a to direct the endonuclease for efficient, RNA-guided DNA cleavage in somatic cells. Key techniques for somatic cell engineering include CRISPR-Cas9 for precise gene edits, such as knockouts, insertions, or corrections, often delivered via or viral vectors in settings. Viral vectors, particularly (AAV), are widely used for gene therapy in somatic cells due to their low , ability to transduce non-dividing cells like neurons and hepatocytes, and capacity to achieve long-term expression without integrating into the host . The first in-human application of CRISPR-based somatic editing occurred in 2016 in , where T cells from a patient were edited to knock out the PD-1 gene, enhancing anti-tumor immunity before reinfusion, marking a milestone in clinical translation. Applications of these methods in focus on correcting monogenic disorders or enhancing immune responses, with modifications being prominent. For instance, CAR-T cell therapy engineers somatic T cells harvested from patients to express chimeric antigen receptors (CARs) via lentiviral or retroviral vectors, redirecting them to target cancer s like in B-cell malignancies, leading to durable remissions in refractory cases. A notable clinical success is Casgevy (exagamglogene autotemcel), approved by the FDA in December 2023 for treating and beta-thalassemia in patients 12 years and older, involving editing of patient-derived hematopoietic stem cells to reactivate production. This approach, approved for leukemias and lymphomas, exemplifies how somatic edits can reprogram immune cells for personalized therapy without affecting transmission. Despite advances, limitations persist, including off-target effects where unintended genomic sites are cleaved, potentially causing mutations or toxicity, as observed in early applications. Immune responses to delivery vectors like AAV or proteins can reduce efficacy and trigger inflammation, necessitating immunosuppressive regimens or engineered hypoimmunogenic variants. Fundamentally, edits are non-heritable, confined to the treated individual and their somatic lineages, avoiding ethical concerns of changes but requiring repeated dosing for proliferative tissues.

Biological Processes

Cell Division and Differentiation

Somatic cells primarily divide through , a process that generates two genetically identical daughter cells, each maintaining the diploid number (2n=46 in humans) to support growth, repair, and maintenance. This division occurs in all non-reproductive cells and is tightly regulated to preserve genomic integrity. The mitotic process unfolds in distinct phases: during , chromosomes condense and the breaks down, while the mitotic begins to form; involves spindle attachment to kinetochores on chromosomes; aligns chromosomes at the cell's equatorial plane; separates toward opposite poles; and reforms nuclear envelopes around the segregated chromosomes. follows, dividing the and organelles to complete the formation of two identical cells. Through these steps, ensures chromosomal stability, with low error rates, approximately 10–20 single variants per in healthy somatic tissues. Following proliferation, somatic cells often undergo , transforming multipotent stem cells into specialized cell types through precise gene regulation that activates tissue-specific programs while silencing others. This process is orchestrated by transcription factors such as , which provide positional information along the body axis and guide segment-specific development in lineages like neurons and muscle cells. For instance, Hox gene clusters are sequentially expressed during embryogenesis to direct the of somatic progenitors into diverse structures, ensuring proper . In most cases, somatic is irreversible, as differentiated cells lose proliferative potential and adopt stable epigenetic modifications, such as and changes, that lock in their functional identity. Cell division and differentiation in somatic cells are governed by regulatory mechanisms, including cell cycle checkpoints that monitor DNA integrity and prevent errors. The G1/S checkpoint, for example, relies on the tumor suppressor to activate p21 and halt progression if DNA damage is detected, thereby safeguarding against propagation of mutations during mitosis. Extracellular signaling pathways further coordinate these events; the Wnt pathway promotes stem cell self-renewal and initiates differentiation in somatic lineages by stabilizing β-catenin for transcriptional activation, while the pathway mediates cell-cell communication to refine fate decisions, such as inhibiting differentiation in progenitors until appropriate cues arise. A prominent example is hematopoiesis, where multipotent hematopoietic s (HSCs), a type of somatic progenitor residing in , differentiate into all lineages—erythrocytes, leukocytes, and platelets—through sequential branching pathways influenced by cytokines and transcription factors like for erythroid commitment. This hierarchical process ensures continuous replenishment of the blood system while maintaining lineage fidelity.

Aging and Senescence

Somatic cells undergo replicative , a process where they cease dividing after a finite number of replications, as first observed in human fibroblasts cultured . This phenomenon, known as the , typically allows approximately 50 divisions before cells enter a permanent state. The primary mechanism driving replicative senescence is shortening, where the protective TTAGGG repeats at ends erode with each due to incomplete . When telomeres become critically short, they are recognized as DNA double-strand breaks, triggering a persistent DNA damage response that activates pathways such as p53/p21 and p16INK4a/, leading to . Additionally, accumulation of other DNA damage, including oxidative lesions and replication , contributes to by amplifying the DNA damage signaling. Cellular senescence in somatic cells manifests in two main types: intrinsic, which is time-dependent and primarily linked to progressive telomere attrition over multiple divisions, and extrinsic, induced by acute stresses such as oxidative damage, irradiation, or oncogenic activation. Intrinsic senescence plays a key role in aging by limiting the regenerative capacity of somatic cell populations, resulting in gradual functional decline across organs. This process has dual implications for organismal : it suppresses tumorigenesis by preventing the of damaged cells, thereby acting as a barrier to cancer development, while its accumulation in tissues promotes age-related pathologies through the (SASP), which fosters chronic inflammation. Notably, most cancer cells evade by reactivating , an enzyme that elongates telomeres and enables indefinite replication.

References

  1. [1]
    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.
  2. [2]
    Definition of somatic cell - NCI Dictionary of Cancer Terms
    Any of the body cells except the reproductive (germ) cells. Search NCI's Dictionary of Cancer Terms. Starts with. Contains. Browse:.
  3. [3]
    The Genome – Introductory Biology: Evolutionary and Ecological ...
    Human body cells (somatic cells) have 46 chromosomes. A somatic cell contains two matched sets of chromosomes, a configuration known as diploid. The letter n is ...
  4. [4]
    Cell Division - Mitosis and Meiosis - Ask A Biologist
    Feb 3, 2014 · Somatic cells make up most of your body's tissues and organs, including skin, muscles, lungs, gut, and hair cells. Reproductive cells (like eggs) ...
  5. [5]
    How do cells divide?: MedlinePlus Genetics
    Mar 26, 2021 · During mitosis, a cell duplicates all of its contents, including its chromosomes, and splits to form two identical daughter cells. Because this ...
  6. [6]
    Meiosis - National Human Genome Research Institute
    In humans, body (or somatic) cells are diploid, containing two sets of chromosomes (one from each parent).Missing: key | Show results with:key
  7. [7]
    Genetics, Somatic Mutation - StatPearls - NCBI Bookshelf - NIH
    Apr 17, 2023 · These mutations do not involve the germline and consequently do not pass on to offspring. Somatic mutations are a normal part of aging and ...
  8. [8]
    Definition of somatic mutation - NCI Dictionary of Cancer Terms
    Somatic mutations can occur in any of the cells of the body except the germ cells (sperm and egg) and therefore are not passed on to children. These alterations ...
  9. [9]
    Stem Cell Basics - University of Nebraska Medical Center
    Somatic cell nuclear transfer (SCNT) is a technique in which the nucleus of a somatic cell, that is any cell of the body apart from the sperm or egg, is ...
  10. [10]
    Somatic Cell Nuclear Transfer in Mammals (1938-2013)
    Nov 4, 2014 · Somatic cells are cells that have gone through the differentiation process and are not germ cells. Somatic cells donate their nuclei, which ...
  11. [11]
    Somatic cells Definition and Examples - Biology Online Dictionary
    May 29, 2023 · The word “somatic” is derived from the Greek word soma, meaning “body”. Hence, all body cells of an organism – apart from the sperm and egg cell ...Missing: etymology | Show results with:etymology
  12. [12]
    August Friedrich Leopold Weismann (1834-1914)
    May 23, 2014 · In this essay, Weismann theorized that the germ-plasm in germ-cells alone, rather than the somatoplasm contained in somatic cells, was ...
  13. [13]
    August Weismann - The Information Philosopher
    Germ cells produce the somatic cells and nothing that changes in the somatic cells can be inherited. Weismann thus denied the Lamarkian theory of evolution, ...
  14. [14]
    Cloning and Stem Cells | Biological Principles
    Cells in an early human embryo, however, are totipotent. Totipotent means they can serve as the basis to form any part of the developing body. Even after ...
  15. [15]
    Glossary - Exploring the Biological Contributions to Human Health
    Somatic cells. Usually any cell of a multicellular organism that will not contribute to the production of gametes, i.e., most cells of which an organism is ...
  16. [16]
    The Origin and Evolution of Cells - The Cell - NCBI Bookshelf - NIH
    Multicellular organisms evolved from unicellular eukaryotes at least 1.7 billion years ago. Some unicellular eukaryotes form multicellular aggregates that ...
  17. [17]
    Volvox, Chlamydomonas, Evolution of Multicellularity - Nature
    The volvocine algae include both unicellular and multicellular organisms that are closely related and exist today (Kirk 1998).
  18. [18]
    Volvox and volvocine green algae | EvoDevo - BioMed Central
    Jul 1, 2020 · Volvocine green algae are a taxonomic group that is uniquely suited to investigating the step-wise acquisition of multicellular organization.
  19. [19]
    Cell differentiation and germ–soma separation in Ediacaran ... - Nature
    Sep 24, 2014 · Cite this article. Chen, L., Xiao, S., Pang, K. et al. Cell differentiation and germ–soma separation in Ediacaran animal embryo-like fossils.
  20. [20]
    Multicellularity makes somatic differentiation evolutionarily stable
    Many multicellular organisms produce two cell lineages: germ cells, whose descendants produce the next generation, and somatic cells, which support, protect, ...Missing: timeline | Show results with:timeline
  21. [21]
    The Evolutionary Origin of Somatic Cells under the Dirty Work ...
    May 13, 2014 · Here we propose the “dirty work hypothesis,” which argues that germ–soma differentiation is an adaptation to allow metabolic work that damages a cell's DNA.Missing: timeline | Show results with:timeline
  22. [22]
    The Germ-Plasm: a Theory of Heredity (1893), by August Weismann
    Jan 26, 2015 · Additionally, Weismann elaborates a theory for variation within a group of related organisms, claiming that variations in germ-plasm cause ...
  23. [23]
    The germ-plasm; a theory of heredity : Weismann, August, 1834-1914
    Feb 11, 2009 · The germ-plasm; a theory of heredity ; Publication date: 1893 ; Topics: Heredity ; Publisher: New York, Scribner's ; Collection: biodiversity; ...
  24. [24]
    What Is Lost in the Weismann Barrier? - PMC - NIH
    Dec 16, 2020 · With his germ plasm theory, Weismann sought to explain how species transform over evolutionary time depending on their environment yet preserve ...
  25. [25]
    Lessons for Inductive Germline Determination - PubMed Central - NIH
    In this review, we compare and contrast these two fundamental mechanisms for germline determination, beginning with the key molecular determinants that play a ...
  26. [26]
    The evolution of insect germline specification strategies - PMC
    In animals, there are two major strategies to specify the germline: maternal provision and zygotic induction. The molecular basis of the maternal provision mode ...
  27. [27]
    Do plants have a segregated germline? - PMC - PubMed Central - NIH
    May 16, 2018 · Germline segregation is the physical separation of the germline from the somatic cell lineages. Segregation is important because once the ...
  28. [28]
    Meiosis and Fertilization - The Cell - NCBI Bookshelf
    Whereas somatic cells undergo mitosis to proliferate, the germ cells undergo meiosis to produce haploid gametes (the sperm and the egg). The development of a ...
  29. [29]
    Cell division: mitosis and meiosis - Biological Principles
    Mitosis produces two daughter cells that are genetically identical to each other, and to the parental cell.
  30. [30]
    Meiosis – Introductory Biology: Evolutionary and Ecological ...
    Somatic cells are sometimes referred to as “body” cells. Homologous chromosomes are matched pairs containing genes for the same traits in identical locations ...18 Meiosis · Meiosis I · Comparing Meiosis And...
  31. [31]
    Chromosomes Fact Sheet
    Aug 15, 2020 · Humans have 23 pairs of chromosomes, for a total of 46 chromosomes. In fact, each species of plants and animals has a set number of chromosomes.Missing: somatic | Show results with:somatic
  32. [32]
    Centromere - National Human Genome Research Institute
    A centromere is a constricted region of a chromosome that separates it into a short arm (p) and a long arm (q).
  33. [33]
    Minute to Understanding: What is a chromosome?
    The tips of the chromosome are capped by sections of DNA called telomeres. Telomeres protect chromosomes during DNA replication. By keeping our DNA secure ...
  34. [34]
    Karyotype - National Human Genome Research Institute
    A karyotype is an individual's complete set of chromosomes. The term also refers to a laboratory-produced image of a person's chromosomes isolated from an ...
  35. [35]
    Polyploidy, the Nucleotype, and Novelty: The Impact of Genome ...
    Polyploidization results in 4n somatic cells; in plants with low chromatin ... Feldman M, AA Levy 2009 Genome evolution in allopolyploid wheat—a revolutionary ...
  36. [36]
    Endoreplication: The Good, the Bad, and the Ugly - PMC - NIH
    Endoreplication is an evolutionarily conserved cell cycle program during which, cells replicate their genomes without division, resulting in polyploid cells.
  37. [37]
    Somatic Mutagenesis in Mammals and Its Implications for Human ...
    Somatic mutations are DNA mutations in somatic tissues, associated with cancer, chronic diseases, and possibly aging, and are not necessary for genetic ...Somatic Mutations: How They... · Somatic Mutation Analysis By... · Aneuploidy And Copy Number...
  38. [38]
    Measuring single cell divisions in human tissues from multi-region ...
    Feb 25, 2020 · We found that mutation rates per cell division were 4–100 times higher in tumours compared to healthy tissue, ranging from 2.91 × 10–9 (bp/ ...
  39. [39]
    Detecting somatic mutations in normal cells - PMC - NIH
    Recent studies have demonstrated that high-throughput sequencing data can be used to detect somatic mutations in non-tumor cells.Missing: consequences | Show results with:consequences
  40. [40]
    Pathogenic mechanisms of somatic mutation and genome ... - NIH
    1. Causes and consequences of somatic DNA mutations. Different types of DNA damage occur daily in every cell of an organism, including strand breaks, base ...
  41. [41]
    Somatic Cell Nuclear Transfer Reprogramming - NIH
    Oct 4, 2019 · Here we summarize recent advances in SCNT technology and its potential applications for both reproductive and therapeutic cloning.Epigenetic Barriers That... · Therapeutic Cloning · Scnt And Ipsc Reprogramming...<|separator|>
  42. [42]
    Adult frogs derived from the nuclei of single somatic cells
    A survey has been made of over 150 adult frogs which have been obtained by the transplantation of nuclei from endoderm cells of Xenopus laevis donors.Missing: paper | Show results with:paper
  43. [43]
    Viable offspring derived from fetal and adult mammalian cells - Nature
    Feb 27, 1997 · We now report the birth of live lambs from three new cell populations established from adult mammary gland, fetus and embryo.
  44. [44]
    A background to nuclear transfer and its applications in agriculture ...
    Cloning animals by nuclear transfer may also be of benefit in conservation and agriculture, for instance: Preservation of genetic diversity and endangered ...
  45. [45]
    Healthy ageing of cloned sheep | Nature Communications
    Jul 26, 2016 · Here we report that SCNT has no obvious detrimental long-term health effects in a cohort of 13 cloned sheep.
  46. [46]
    Epigenetic abnormalities associated with somatic cell nuclear ...
    Jun 11, 2021 · Moreover, injection of Kdm4d and Kdm5b together synergistically improved the SCNT efficiency and over 95% of reconstructed embryos successfully ...
  47. [47]
    Biobanking and Regenerative Medicine: An Overview - PMC - NIH
    May 31, 2018 · This Special Issue will address the topic of biobanking and how it fits into regenerative medicine. Topics such as stem cell banking (e.g., cord ...
  48. [48]
    Dimethyl sulfoxide-free cryopreservation for cell therapy: A review
    Dimethyl sulfoxide (Me2SO) has been the cryoprotectant of choice in most biobanking situations due to its exceptional performance in mitigating freezing-related ...
  49. [49]
    Increased Cell Survival of Human Primary Conjunctival Stem Cells ...
    DMSO is the standard cryopreservation reagent for biobanking several human cell types, including several pluripotent stem cells and progenitor stem cells.
  50. [50]
    Chemical approaches to cryopreservation | Nature Reviews Chemistry
    Jul 18, 2022 · Stocks of red blood cells are cryopreserved using 20–40 wt% glycerol, immortalized cell lines are routinely stored in 10% (v/v) DMSO and cells ...
  51. [51]
    Human iPSC banking: barriers and opportunities
    Oct 28, 2019 · The introduction of induced pluripotent stem cells (iPSCs) has opened up the potential for personalized cell therapies and ushered in new ...
  52. [52]
    Umbilical cord blood and cord tissue banking as somatic stem cell ...
    Dec 5, 2023 · Human umbilical cord blood (CB) and umbilical cord tissue (UC) are attractive sources of somatic stem cells for gene and cell therapies.
  53. [53]
    Cord Blood vs. Cord Tissue: Differences, & Benefits of Each
    Apr 22, 2024 · Cord blood is rich in Hematopoietic Stem Cells (HSCs), while cord tissue contains Mesenchymal Stem Cells (MSCs). Both types of stem cells are very valuable.Missing: biobanks HeLa somatic
  54. [54]
    What Role do Biobanks Play in Cancer Research? - News-Medical
    Mar 24, 2023 · Long before then, cell line biobanking can be traced back to the 1950s and the Johns Hopkins Hospital in Maryland. Here, cancer cells were ...Missing: somatic | Show results with:somatic
  55. [55]
    Our history - UK Biobank
    Oct 2, 2025 · UK Biobank was established in the early 2000s to enable researchers around the world to make discoveries that improve human health.<|separator|>
  56. [56]
    Ethical aspects of human biobanks: a systematic review - PMC - NIH
    Ethical issues are commonly present in many aspects of biobanking. The fact that biobanks deal with human samples, invading an individual autonomy or limiting ...
  57. [57]
    Ethical considerations for biobanks serving underrepresented ... - NIH
    Dec 10, 2024 · Participant privacy is a major ethical concern in biobanking, as the collected biospecimens and data often contain sensitive personal ...2. Ethical Considerations... · 4. Case Studies Of Biobanks... · Footnotes
  58. [58]
    Quality management, ethical considerations, and emerging ...
    Mar 1, 2025 · The integration of genomics and biobanking promises to increase the effectiveness and precision of personalized medicine. This article ...Introduction · Genomic Medicine · Biobank-Related Challenges
  59. [59]
    Genome Engineering With Zinc-Finger Nucleases - PMC - NIH
    The first ZFNs were created as chimeric restriction endonucleases and were shown to have in vitro activity (Kim et al. 1996). It was not clear that the ...
  60. [60]
    Efficient design and assembly of custom TALEN and other TAL ...
    Abstract. TALENs are important new tools for genome engineering. Fusions of transcription activator-like (TAL) effectors of plant pathogenic Xanthomonas sp.Materials And Methods · Results · Targeted Mutagenesis In...
  61. [61]
    A Programmable Dual-RNA–Guided DNA Endonuclease ... - Science
    Jun 28, 2012 · Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA- ...
  62. [62]
    Somatic Genome Editing - NCBI
    The use of human genome editing to make edits in somatic cells for purposes of treating genetically inherited diseases is already in clinical trials.HOMOLOGOUS AND... · POTENTIAL HUMAN... · SCIENTIFIC AND TECHNICAL...
  63. [63]
    Adeno-Associated Virus (AAV) as a Vector for Gene Therapy - PMC
    This review will provide an overview of some important factors to consider in the use of AAV as a vector for gene therapy.
  64. [64]
    CRISPR gene-editing tested in a person for the first time - Nature
    Nov 15, 2016 · A Chinese group has become the first to inject a person with cells that contain genes edited using the revolutionary CRISPR–Cas9 technique.
  65. [65]
    CAR-T cell therapy for cancer: current challenges and future directions
    Jul 4, 2025 · In a landmark 2003 study, the MSKCC team demonstrated the feasibility of expanding CAR-T cells ex vivo using artificial antigen-presenting cells ...
  66. [66]
    Off-target effects in CRISPR/Cas9 gene editing - PMC - NIH
    They found that the number of off-target sites reduced by 98.7%, 95.4% and 94.1%, respectively. Meanwhile, the absolute on-target activity of these Cas9 mutants ...Missing: heritable | Show results with:heritable
  67. [67]
  68. [68]
    Phases of mitosis | Mitosis | Biology (article) - Khan Academy
    How a cell divides to make two genetically identical cells. Prophase, metaphase, anaphase, and telophase.
  69. [69]
    Mitosis - PMC - PubMed Central - NIH
    Each step of the mitotic process is illuminated by discoveries in a range of research fields and organisms.
  70. [70]
    Mitosis - National Human Genome Research Institute
    Mitosis is the process by which a cell replicates its chromosomes and then segregates them, producing two identical nuclei in preparation for cell division.
  71. [71]
    Hox genes in development and beyond
    Jan 16, 2023 · Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms.Initiation And Regulation Of... · Regional Specificity And... · Continuing Roles For Hox...
  72. [72]
    Role of Hox genes in stem cell differentiation - PMC - PubMed Central
    Hox genes are an evolutionary highly conserved gene family. They determine the anterior-posterior body axis in bilateral organisms and influence the ...
  73. [73]
    HOX genes in stem cells: Maintaining cellular identity and regulation ...
    Understanding the mechanism of HOX gene regulation in stem cells may provide new ways to manipulate stem cell fate and function leading to improved and new ...Abstract · Introduction · Conclusion
  74. [74]
    DNA damage triggers a prolonged p53-dependent G1 arrest and ...
    Abstract. The tumor suppressor p53 is a cell cycle checkpoint protein that contributes to the preservation of genetic stability by mediating either a G1 arrest ...
  75. [75]
    Wnt/Notch signalling and information processing during development
    Feb 1, 2008 · The Wnt and Notch signalling pathways represent two major channels of communication used by animal cells to control their identities and behaviour during ...
  76. [76]
    Stem Cells, Hematopoiesis and Lineage Tracing - NIH
    Apr 27, 2022 · Hematopoiesis refers to the highly regulated processes in which individual blood cells are formed. In steady state, the different blood cell ...
  77. [77]
    Signaling pathways governing stem-cell fate - ASH Publications
    Several developmentally conserved signaling pathways have emerged as important control devices of HSC fate, including Notch, Wingless-type (Wnt), Sonic ...
  78. [78]
    The serial cultivation of human diploid cell strains - ScienceDirect
    December 1961, Pages 585-621. Experimental Cell Research. The serial cultivation of human diploid cell strains☆. Author links open overlay panel. L. Hayflick ,Missing: original | Show results with:original
  79. [79]
    Telomeres shorten during ageing of human fibroblasts - Nature
    May 31, 1990 · Here, we show that the amount and length of telomeric DNA in human fibroblasts does in fact decrease as a function of serial passage during ageing in vitro and ...
  80. [80]
    Human cell senescence as a DNA damage response - ScienceDirect
    The DNA damage response pathway triggers replicative senescence. In recent years, the early events in DNA damage-induced signalling have been very well ...Telomeres And Replicative... · Telomere-Dependent... · The Dna Damage Response...
  81. [81]
    Mechanisms and functions of cellular senescence - JCI
    Apr 2, 2018 · Currently, senescence is considered a stress response that can be induced by a wide range of intrinsic and extrinsic insults, including ...
  82. [82]
    Senescence and immortalization: role of telomeres and telomerase
    Most malignant tumors must have a mechanism for bypassing senescence to have the unlimited proliferative capacity that appears to be required for advanced ...