Human somatic variation

Last updated

Human somatic variations are somatic mutations (mutations that occur in somatic cells) both at early stages of development and in adult cells. These variations can lead either to pathogenic phenotypes or not, even if their function in healthy conditions is not completely clear yet. [1]

Contents

The term mosaic (from medieval Latin musaicum, meaning "work of the Muses") [2] has been used since antiquity to refer to an artistic patchwork of ornamental stones, glass, gems, or other precious material. At a distance, the collective image appears as it would in a painting; only on close inspection do the individual components become recognizable. [2] In biological systems, mosaicism implies the presence of more than one genetically distinct cell line in a single organism. Occurrence of this phenomenon not only can result in major phenotypic changes but also reveal the expression of otherwise lethal genetic mutations. [3]

Genetic mutations involved in mosaicism may be due to endogenous factors, such as transposons and ploidy changes, or exogenous factors, such as UV radiation and nicotine. [4]

Somatic mosaicism in healthy human tissues

Somatic mosaicism arises a result of somatic mutations: genomic (or even mitochondrial) alterations of different sizes ranging from a single nucleotide to chromosome gains or loss within somatic cells. These alterations within somatic cells begin at an early stage (pre-implantation or conception) and continue during aging, giving rise to phenotypic heterogeneity within cells, which may lead to the development of diseases such as cancer. [4] Novel array based techniques for screening genome-wide copy number variants and loss of heterozygosity in single cells showed that chromosome aneuploidies, uniparental disomies, segmental deletions, duplications, and amplifications frequently occur during embryogenesis. [5] Yet not all somatic mutations are propagated to the adult individual, due to the phenomenon of cell competition. [6]

Genetic alterations involving gains or loss of entire chromosomes predominantly occur during anaphase stage of cell division. But these are uncommon in somatic cells because they are usually selected against due to their deleterious consequences. [7] Somatic variations during embryonic development can be represented by monozygous twins since they carry different copy number profiles and epigenetic marks that keep on increasing with age. [8]

Early research on somatic mutations in aging showed that deletions, inversion, and translocations of genetic material are common in aging mice and aging genomes tend to contain visible chromosomal changes, mitotic recombination, whole gene deletions, intragenic deletions, and point mutations. Other factors include the loss of methylation, increasing gene expression heterogeneity correlating to genomic abnormalities, [4] and telomere shortening. [9] It is uncertain if transcription-based DNA repair takes part in the maintaining of somatic mutations in aging tissues. [4]

In some cells, the somatically acquired alterations can be reversed back to wild type alleles by reversion mosaicism. This can be due to endogenous mechanism such as homologous recombination, codon substitution, second-site suppressor mutations, DNA slippage, and mobile elements. [10]

Somatic cancer-associated mutations in normal tissues

The advent of Next-Generation Sequencing technologies has increased the resolution of mutation detection and has led to the revelation that older individuals not only accumulate chromosomal alterations but also abundant mutations in cancer driver genes. [11]

Age-associated accumulation of chromosomal alterations has been documented with a variety of cytogenetic approaches, from chromosome painting to single nucleotide polymorphism (SNP) arrays. [12]

Numerous studies demonstrated that the clonal populations might lead to loss of organismal health through the functional decline of tissue and/or the promotion of disease processes, such as cancer. This is the reason why the aberrant clonal expansion (ACE) resulting from cancer-associated mutations are common in noncancerous tissue and accumulate with age. This is universal in most organisms and affects multiple tissues. [11]

In the hematopoietic compartment mutations include both large structural chromosomal alterations and point mutations affecting cancer-associated genes. Some translocations appear to occur very early in life. The frequency of these events is low in people younger than 50 years (<0.5%), but this frequency rapidly increases to 2% to 3% of individuals in their 70s and 80s. This phenomenon was termed clonal hematopoiesis. A number of environmental factors, such as smoking, viral infections, and pesticide exposure, may contribute not only through mutation induction but also by modulation of clonal expansion. [13]

Otherwise, the detection of somatic variants in normal solid tissues has historically proved difficult. The main reasons are the generally slower replicative index, clonally restrictive tissue architecture, difficulty of tissue access, and low frequency of mutation occurrence. Recently, the analysis of somatic mutations in benign tissues adjacent to tumors revealed that 80% of samples harbors clonal mutations, with increased frequency associated with older age, smoking, and concurrent mutations in DNA repair genes. With the advent of NGS, it has become increasingly clear that somatic mutations accumulate with aging in normal tissue, even in individuals who are cancer-free. [11]

This suggested that clonal expansions driven by cancer genes are a near-universal feature of aging. NGS technologies revealed that the clonal expansions of cancer-associated mutations are very common condition in somatic tissues. [11]

Human somatic variations in brain

Through several recent studies a prevalence of somatic variations, both in pathological and healthy nervous systems, has been highlighted. [14] [15]

Somatic aneuploidy such as SNVs (single-nucleotide variations) and CNVs (copy number variations) have been particularly observed and linked to brain disfunctions when arising in prenatal brain development; anyway those somatic aneuploidy have been observed in rates of 1,3-40%, potentially increasing with age and for this reason they have been proposed as a mechanism to generate normal genetic diversity among neurons. [16]

The confirmation of that hypothesis has been obtained through studies of single-cell sequencing, which allow a direct assessment of single neuronal genomes, so that a systematic characterization of somatic aneuploidies and subchromosomal CNVs of these cells is possible. Using postmortem brains of both healthy and diseased humans it has been possible to study how CNVs change among these two groups. It emerged that somatic aneuploidies in healthy brains are quite rare, but somatic CNVs instead aren't. [17]

These studies also showed that clonal CNVs exist in both pathological and healthy brains. This means that some CNVs can arise in early development without causing diseases, even though, when compared to the CNVs arising in other cell types such as lymphoblast, the brain's ones are more often private. This evidence could be given by the fact that, while lymphoblasts can generate clonal CNVs for a long period as they continue to proliferate, adult neurons do not replicate anymore, so the clonal CNVs they are carrying must have been generated in an early development stage. [17]

Data highlighted a tendency in neurons for the loss, rather than for the gain of copies when compared to lymphoblasts. These differences could suggest that the molecular mechanisms of CNVs arising in that two cell types are completely different. [17]

L1-associated mosaicism in brain cells

The retrotransposon LINE-1 (long interspersed element 1, L1) is a transposable element that has colonized the mammalian germline. L1 retrotransposition can happen also in somatic cells causing mosaicism (SLAVs – L1-associated variations) and in cancer. Retrotransposition is a copy and paste process in which the RNA template is retrotranscribed in DNA and integrated randomly in the genome. In humans there are around 500.000 copies of L1 and occupy 17% of genome. Its mRNA encodes for two proteins; one of them in particular has a reverse transcriptase and endonuclease activity that allows the retrotransposition in cis. Anyway most part of these copies are rendered immobile by mutations or 5’ truncation, leaving just about 80–100 mobile L1 per human genome and just about 10 are considered hot L1s so able to mobilize efficiently. [18]

L1 transpose using a mechanism called TPRT (target primed reverse transcription) it's able to insert a L1 endonuclease motif, target site duplications (TSD) and a poly-A tail with a cis preference. [19]

It has been seen in the past that there's L1 mobilization in neural progenitors during foetal and adult neurogenesis suggesting that the brain may be a L1 mosaicism hotspot. Moreover, some studies suggested that also non-dividing neurons can support L1 mobilization. This has been confirmed by single-cell genomic studies. [18]

Single-cell paired-end sequencing experiments found out that SLAVs are present both in neurons and glia of hippocampus and frontal cortex. Any neural cell has a similar probability to contain a SLAV, suggesting that somatic variations are a random phaenomenon, not focused on a specific group of cells. SLAVs occurrence in the brain is estimated to be of 0.58–1 SLAVs per cell and to involve 44–63% of the brain cells.[ citation needed ]  

Since experiments showed that a half of the analyzed SLAVs lack target site duplication (TSD), another kind of L1-associated variant might occur. In fact those sequences don't have an endonuclease activity, but still have endonuclease motifs so that they can be retrotransposed in trans.[ citation needed ]

An application of the study of somatic mosaicism in the brain could be the tracing of specific brain cells. Indeed, if the somatic L1 insertions occurs in a progenitor cell, the unique variant could be used to trace the progenitor cell's development, localization, and spreading through the brain. On the contrary, if the somatic L1 insertion occurs late in development, it will be present just in a single cell or in a small group of cells. Therefore, tracing somatic variations could be useful to understand at which point of development they have occurred. Further experiments are necessary to understand the role of somatic mosaicism in brain function, since small groups of cells or even single cells can affect network activity. [20]

Human somatic variations and the immune system

Human somatic mutations (HSMs) are intensively exploited by the immune system for the production of antibodies. HSMs, recombination in particular, are indeed the reason why antibodies can identify an epitope with such high specificity and sensitivity. [21]

Antibodies are encoded by B cells. Each antibody is composed of two heavy chains (IgH, encoded by IGH gene) and two light chains (IgL, encoded by either IGL or IGK gene). Each chain is then composed of a constant region (C) and a variable region (V). The constant region (C) on the heavy chain is important in the BCR signaling and determines the type of immunoglobuline (IgA, IgD, IgE, IgG, or IgM). The variable region (V) is responsible for the recognition of the target epitope and is the product of recombination processes in the related loci. [22]

After exposure of an antigen, B cells start developing. B cells genome undergoes repeated recombination processing on the Ig genes until the recognition of the epitope is perfectioned. The recombination involves the IGH locus first and then the IGL and IGK loci. All IGL, IGK, and IGH genes are the product of the V(D)J recombination process. This recombination involves the variable (V), diversity (D) and joining (J) segments. All three segments (V, D, J) are involved in the formation of the heavy chain, while only V and J recombination products encode for the light chain. [23]

The recombination between these regions allows the formation of 1012–1018 potential different sequences. However, this number is an overestimation, since many factors contribute to limit the diversity of the B cell repertoire, first of all the actual number of B cell in the organism. [23]

Cardiac mosaicism

Somatic mosaicism has been noted in the heart. Sequencing suggested mosaic variation in the gap junction protein connexin in three patients out of 15 might contribute to atrial fibrillation [24] although subsequent reports in larger numbers of patients found no examples among a large panel of genes. [25] At Stanford, a team led by Euan Ashley demonstrated somatic mosaicism in the heart of a newborn presenting with life threatening arrhythmia. Family-based genome sequencing as well as tissue RNA sequencing and single cell genomics techniques were used to verify the finding. A model combining partial and ordinary differential equations with inputs from heterologous single channel electrophysiology experiments of the genetic variant recapitulated certain aspects of the clinical presentation. [26]

See also

Related Research Articles

<span class="mw-page-title-main">Mutation</span> Alteration in the nucleotide sequence of a genome

In biology, a mutation is an alteration in the nucleic acid sequence of the genome of an organism, virus, or extrachromosomal DNA. Viral genomes contain either DNA or RNA. Mutations result from errors during DNA or viral replication, mitosis, or meiosis or other types of damage to DNA, which then may undergo error-prone repair, cause an error during other forms of repair, or cause an error during replication. Mutations may also result from insertion or deletion of segments of DNA due to mobile genetic elements.

<span class="mw-page-title-main">Aneuploidy</span> Presence of an abnormal number of chromosomes in a cell

Aneuploidy is the presence of an abnormal number of chromosomes in a cell, for example a human cell having 45 or 47 chromosomes instead of the usual 46. It does not include a difference of one or more complete sets of chromosomes. A cell with any number of complete chromosome sets is called a euploid cell.

A genetic screen or mutagenesis screen is an experimental technique used to identify and select individuals who possess a phenotype of interest in a mutagenized population. Hence a genetic screen is a type of phenotypic screen. Genetic screens can provide important information on gene function as well as the molecular events that underlie a biological process or pathway. While genome projects have identified an extensive inventory of genes in many different organisms, genetic screens can provide valuable insight as to how those genes function.

Gene duplication is a major mechanism through which new genetic material is generated during molecular evolution. It can be defined as any duplication of a region of DNA that contains a gene. Gene duplications can arise as products of several types of errors in DNA replication and repair machinery as well as through fortuitous capture by selfish genetic elements. Common sources of gene duplications include ectopic recombination, retrotransposition event, aneuploidy, polyploidy, and replication slippage.

<span class="mw-page-title-main">Mosaic (genetics)</span> Condition in multi-cellular organisms

Mosaicism or genetic mosaicism is a condition in which a multicellular organism possesses more than one genetic line as the result of genetic mutation. This means that various genetic lines resulted from a single fertilized egg. Mosaicism is one of several possible causes of chimerism, wherein a single organism is composed of cells with more than one distinct genotype.

<span class="mw-page-title-main">Germline mutation</span> Inherited genetic variation

A germline mutation, or germinal mutation, is any detectable variation within germ cells. Mutations in these cells are the only mutations that can be passed on to offspring, when either a mutated sperm or oocyte come together to form a zygote. After this fertilization event occurs, germ cells divide rapidly to produce all of the cells in the body, causing this mutation to be present in every somatic and germline cell in the offspring; this is also known as a constitutional mutation. Germline mutation is distinct from somatic mutation.

Heteroplasmy is the presence of more than one type of organellar genome within a cell or individual. It is an important factor in considering the severity of mitochondrial diseases. Because most eukaryotic cells contain many hundreds of mitochondria with hundreds of copies of mitochondrial DNA, it is common for mutations to affect only some mitochondria, leaving most unaffected.

Genetics, a discipline of biology, is the science of heredity and variation in living organisms.

<span class="mw-page-title-main">L1 (protein)</span> Mammalian protein found in Homo sapiens

L1, also known as L1CAM, is a transmembrane protein member of the L1 protein family, encoded by the L1CAM gene. This protein, of 200-220 kDa, is a neuronal cell adhesion molecule with a strong implication in cell migration, adhesion, neurite outgrowth, myelination and neuronal differentiation. It also plays a key role in treatment-resistant cancers due to its function. It was first identified in 1984 by M. Schachner who found the protein in post-mitotic mice neurons.

Carcinogenesis, also called oncogenesis or tumorigenesis, is the formation of a cancer, whereby normal cells are transformed into cancer cells. The process is characterized by changes at the cellular, genetic, and epigenetic levels and abnormal cell division. Cell division is a physiological process that occurs in almost all tissues and under a variety of circumstances. Normally, the balance between proliferation and programmed cell death, in the form of apoptosis, is maintained to ensure the integrity of tissues and organs. According to the prevailing accepted theory of carcinogenesis, the somatic mutation theory, mutations in DNA and epimutations that lead to cancer disrupt these orderly processes by interfering with the programming regulating the processes, upsetting the normal balance between proliferation and cell death. This results in uncontrolled cell division and the evolution of those cells by natural selection in the body. Only certain mutations lead to cancer whereas the majority of mutations do not.

<span class="mw-page-title-main">Copy number variation</span> Repeated DNA variation between individuals

Copy number variation (CNV) is a phenomenon in which sections of the genome are repeated and the number of repeats in the genome varies between individuals. Copy number variation is a type of structural variation: specifically, it is a type of duplication or deletion event that affects a considerable number of base pairs. Approximately two-thirds of the entire human genome may be composed of repeats and 4.8–9.5% of the human genome can be classified as copy number variations. In mammals, copy number variations play an important role in generating necessary variation in the population as well as disease phenotype.

Mitotic recombination is a type of genetic recombination that may occur in somatic cells during their preparation for mitosis in both sexual and asexual organisms. In asexual organisms, the study of mitotic recombination is one way to understand genetic linkage because it is the only source of recombination within an individual. Additionally, mitotic recombination can result in the expression of recessive genes in an otherwise heterozygous individual. This expression has important implications for the study of tumorigenesis and lethal recessive genes. Mitotic homologous recombination occurs mainly between sister chromatids subsequent to replication. Inter-sister homologous recombination is ordinarily genetically silent. During mitosis the incidence of recombination between non-sister homologous chromatids is only about 1% of that between sister chromatids.

Somatic evolution is the accumulation of mutations and epimutations in somatic cells during a lifetime, and the effects of those mutations and epimutations on the fitness of those cells. This evolutionary process has first been shown by the studies of Bert Vogelstein in colon cancer. Somatic evolution is important in the process of aging as well as the development of some diseases, including cancer.

Cancer genome sequencing is the whole genome sequencing of a single, homogeneous or heterogeneous group of cancer cells. It is a biochemical laboratory method for the characterization and identification of the DNA or RNA sequences of cancer cell(s).

Genome instability refers to a high frequency of mutations within the genome of a cellular lineage. These mutations can include changes in nucleic acid sequences, chromosomal rearrangements or aneuploidy. Genome instability does occur in bacteria. In multicellular organisms genome instability is central to carcinogenesis, and in humans it is also a factor in some neurodegenerative diseases such as amyotrophic lateral sclerosis or the neuromuscular disease myotonic dystrophy.

Multiple Annealing and Looping Based Amplification Cycles (MALBAC) is a quasilinear whole genome amplification method. Unlike conventional DNA amplification methods that are non-linear or exponential, MALBAC utilizes special primers that allow amplicons to have complementary ends and therefore to loop, preventing DNA from being copied exponentially. This results in amplification of only the original genomic DNA and therefore reduces amplification bias. MALBAC is “used to create overlapped shotgun amplicons covering most of the genome”. For next generation sequencing, MALBAC is followed by regular PCR which is used to further amplify amplicons.

Tumour heterogeneity describes the observation that different tumour cells can show distinct morphological and phenotypic profiles, including cellular morphology, gene expression, metabolism, motility, proliferation, and metastatic potential. This phenomenon occurs both between tumours and within tumours. A minimal level of intra-tumour heterogeneity is a simple consequence of the imperfection of DNA replication: whenever a cell divides, a few mutations are acquired—leading to a diverse population of cancer cells. The heterogeneity of cancer cells introduces significant challenges in designing effective treatment strategies. However, research into understanding and characterizing heterogeneity can allow for a better understanding of the causes and progression of disease. In turn, this has the potential to guide the creation of more refined treatment strategies that incorporate knowledge of heterogeneity to yield higher efficacy.

<span class="mw-page-title-main">LINE1</span> Group of transposable elements

LINE1 is a family of related class I transposable elements in the DNA of some organisms, classified with the long interspersed nuclear elements (LINEs). L1 transposons comprise approximately 17% of the human genome. These active L1s can interrupt the genome through insertions, deletions, rearrangements, and copy number variations. L1 activity has contributed to the instability and evolution of genomes and is tightly regulated in the germline by DNA methylation, histone modifications, and piRNA. L1s can further impact genome variation through mispairing and unequal crossing over during meiosis due to its repetitive DNA sequences.

PyClone is a software that implements a Hierarchical Bayes statistical model to estimate cellular frequency patterns of mutations in a population of cancer cells using observed alternate allele frequencies, copy number, and loss of heterozygosity (LOH) information. PyClone outputs clusters of variants based on calculated cellular frequencies of mutations.

A somatic mutation is a change in the DNA sequence of a somatic cell of a multicellular organism with dedicated reproductive cells; that is, any mutation that occurs in a cell other than a gamete, germ cell, or gametocyte. Unlike germline mutations, which can be passed on to the descendants of an organism, somatic mutations are not usually transmitted to descendants. This distinction is blurred in plants, which lack a dedicated germline, and in those animals that can reproduce asexually through mechanisms such as budding, as in members of the cnidarian genus Hydra.

References

  1. Li, Chun; Williams, Scott M. (2013-12-01). "Human Somatic Variation: It's Not Just for Cancer Anymore". Current Genetic Medicine Reports. 1 (4): 212–218. doi: 10.1007/s40142-013-0029-z . ISSN   2167-4876.
  2. 1 2 "mosaic | Origin and meaning of mosaic by Online Etymology Dictionary". etymonline.com. Retrieved 2019-11-21.
  3. Youssoufian, Hagop; Pyeritz, Reed E. (October 2002). "Mechanisms and consequences of somatic mosaicism in humans". Nature Reviews Genetics. 3 (10): 748–758. doi:10.1038/nrg906. ISSN   1471-0064. PMID   12360233. S2CID   6355589.
  4. 1 2 3 4 De, Subhajyoti (2011-06-01). "Somatic mosaicism in healthy human tissues". Trends in Genetics. 27 (6): 217–223. doi:10.1016/j.tig.2011.03.002. ISSN   0168-9525. PMID   21496937.
  5. Vanneste, Evelyne; Voet, Thierry; Le Caignec, Cédric; Ampe, Michèle; Konings, Peter; Melotte, Cindy; Debrock, Sophie; Amyere, Mustapha; Vikkula, Miikka; Schuit, Frans; Fryns, Jean-Pierre (May 2002). "Chromosome instability is common in human cleavage-stage embryos" (PDF). Nature Medicine. 15 (5): 577–583. doi:10.1038/nm.1924. ISSN   1546-170X. PMID   19396175. S2CID   10530085.
  6. Kalousek, D. K.; Dill, F. J. (1983-08-12). "Chromosomal mosaicism confined to the placenta in human conceptions". Science. 221 (4611): 665–667. Bibcode:1983Sci...221..665K. doi:10.1126/science.6867735. ISSN   0036-8075. PMID   6867735.
  7. Leach, Natalia T.; Rehder, Catherine; Jensen, Keith; Holt, Shawn; Jackson-Cook, Colleen (2004-08-01). "Human chromosomes with shorter telomeres and large heterochromatin regions have a higher frequency of acquired somatic cell aneuploidy". Mechanisms of Ageing and Development. 125 (8): 563–573. doi:10.1016/j.mad.2004.06.006. ISSN   0047-6374. PMID   15336914. S2CID   19899116.
  8. Bruder, Carl E. G.; Piotrowski, Arkadiusz; Gijsbers, Antoinet A. C. J.; Andersson, Robin; Erickson, Stephen; Diaz de Ståhl, Teresita; Menzel, Uwe; Sandgren, Johanna; von Tell, Desiree; Poplawski, Andrzej; Crowley, Michael (2008-03-03). "Phenotypically Concordant and Discordant Monozygotic Twins Display Different DNA Copy-Number-Variation Profiles". The American Journal of Human Genetics. 82 (3): 763–771. doi:10.1016/j.ajhg.2007.12.011. ISSN   0002-9297. PMC   2427204 . PMID   18304490.
  9. Halaschek-Wiener, Julius; Vulto, Irma; Fornika, Dan; Collins, Jennifer; Connors, Joseph M.; Le, Nhu D.; Lansdorp, Peter M.; Brooks-Wilson, Angela (2008-11-01). "Reduced telomere length variation in healthy oldest old". Mechanisms of Ageing and Development. 129 (11): 638–641. doi:10.1016/j.mad.2008.07.004. ISSN   0047-6374. PMID   18765247. S2CID   2595117.
  10. Ellis, N A; Lennon, D J; Proytcheva, M; Alhadeff, B; Henderson, E E; German, J (November 1995). "Somatic intragenic recombination within the mutated locus BLM can correct the high sister-chromatid exchange phenotype of Bloom syndrome cells". American Journal of Human Genetics. 57 (5): 1019–1027. ISSN   0002-9297. PMC   1801389 . PMID   7485150.
  11. 1 2 3 4 Risques, Rosa Ana; Kennedy, Scott R. (2018-01-04). "Aging and the rise of somatic cancer-associated mutations in normal tissues". PLOS Genetics. 14 (1): e1007108. doi: 10.1371/journal.pgen.1007108 . ISSN   1553-7404. PMC   5754046 . PMID   29300727.
  12. Jacobs, Kevin B; Yeager, Meredith; Zhou, Weiyin; Wacholder, Sholom; Wang, Zhaoming; Rodriguez-Santiago, Benjamin; Hutchinson, Amy; Deng, Xiang; Liu, Chenwei; Horner, Marie-Josephe; Cullen, Michael (June 2012). "Detectable clonal mosaicism and its relationship to aging and cancer". Nature Genetics. 44 (6): 651–658. doi:10.1038/ng.2270. ISSN   1061-4036. PMC   3372921 . PMID   22561519.
  13. Machiela, Mitchell J.; Zhou, Weiyin; Yeager, Meredith; Jacobs, Kevin; Berndt, Sonja; Lan, Qing; Rothman, Nathaniel; Savage, Sharon; Taylor, Phil R.; deVivo, Immaculata; Hoover, Robert N. (2013-04-15). "Abstract 2542: Evidence for large somatic structural events detected as autosomal genetic mosaicism in GWAS data.". Epidemiology. p. 2542. doi:10.1158/1538-7445.am2013-2542.{{cite book}}: |journal= ignored (help)
  14. Biesecker, Leslie G.; Spinner, Nancy B. (May 2013). "A genomic view of mosaicism and human disease". Nature Reviews Genetics. 14 (5): 307–320. doi:10.1038/nrg3424. ISSN   1471-0064. PMID   23594909. S2CID   4494815.
  15. Poduri, Annapurna; Evrony, Gilad D.; Cai, Xuyu; Walsh, Christopher A. (2013-07-05). "Somatic Mutation, Genomic Variation, and Neurological Disease". Science. 341 (6141): 1237758. doi:10.1126/science.1237758. ISSN   0036-8075. PMC   3909954 . PMID   23828942.
  16. Bushman, Diane M.; Chun, Jerold (2013-04-01). "The genomically mosaic brain: Aneuploidy and more in neural diversity and disease". Seminars in Cell & Developmental Biology. WASP/WAVE proteins: expanding members and functions & The role of ploidy variation on cellular adaptation. 24 (4): 357–369. doi:10.1016/j.semcdb.2013.02.003. ISSN   1084-9521. PMC   3637860 . PMID   23466288.
  17. 1 2 3 Cai, Xuyu; Evrony, Gilad D.; Lehmann, Hillel S.; Elhosary, Princess C.; Mehta, Bhaven K.; Poduri, Annapurna; Walsh, Christopher A. (2014-09-11). "Single-Cell, Genome-wide Sequencing Identifies Clonal Somatic Copy-Number Variation in the Human Brain". Cell Reports. 8 (5): 1280–1289. doi:10.1016/j.celrep.2014.07.043. ISSN   2211-1247. PMC   4272008 . PMID   25159146.
  18. 1 2 Faulkner, Geoffrey J.; Garcia-Perez, Jose L. (November 2017). "L1 Mosaicism in Mammals: Extent, Effects, and Evolution". Trends in Genetics. 33 (11): 802–816. doi: 10.1016/j.tig.2017.07.004 . ISSN   0168-9525. PMID   28797643.
  19. Luan, D. D.; Korman, M. H.; Jakubczak, J. L.; Eickbush, T. H. (1993-02-26). "Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition". Cell. 72 (4): 595–605. doi:10.1016/0092-8674(93)90078-5. ISSN   0092-8674. PMID   7679954. S2CID   42587840.
  20. Aimone, J. B.; Deng, W.; Gage, F. H. (2010). "Adult neurogenesis: integrating theories and separating functions". Trends in Cognitive Sciences. 14 (7): 325–337. doi:10.1016/j.tics.2010.04.003. PMC   2904863 . PMID   20471301.
  21. Wardemann, Hedda; Busse, Christian E. (2017-07-01). "Novel Approaches to Analyze Immunoglobulin Repertoires". Trends in Immunology. 38 (7): 471–482. doi:10.1016/j.it.2017.05.003. ISSN   1471-4906. PMID   28566130.
  22. Padlan, E. A. (February 1994). "Anatomy of the antibody molecule". Molecular Immunology. 31 (3): 169–217. doi:10.1016/0161-5890(94)90001-9. ISSN   0161-5890. PMID   8114766.
  23. 1 2 Imkeller, Katharina; Wardemann, Hedda (2018). "Assessing human B cell repertoire diversity and convergence". Immunological Reviews. 284 (1): 51–66. doi: 10.1111/imr.12670 . ISSN   1600-065X. PMID   29944762.
  24. Gollob, Michael H.; Jones, Douglas L.; Krahn, Andrew D.; Danis, Lynne; Gong, Xiang-Qun; Shao, Qing; Liu, Xiaoqin; Veinot, John P.; Tang, Anthony S.L.; Stewart, Alexandre F.R.; Tesson, Frederique; Klein, George J.; Yee, Raymond; Skanes, Allan C.; Guiraudon, Gerard M.; Ebihara, Lisa; Bai, Donglin (22 June 2006). "Somatic Mutations in the Connexin 40 Gene ( GJA5 ) in Atrial Fibrillation". New England Journal of Medicine. 354 (25): 2677–2688. doi:10.1056/NEJMoa052800. PMID   16790700.
  25. Roberts, Jason D.; Longoria, James; Poon, Annie; Gollob, Michael H.; Dewland, Thomas A.; Kwok, Pui-Yan; Olgin, Jeffrey E.; Deo, Rahul C.; Marcus, Gregory M. (February 2015). "Targeted Deep Sequencing Reveals No Definitive Evidence for Somatic Mosaicism in Atrial Fibrillation". Circulation: Cardiovascular Genetics. 8 (1): 50–57. doi: 10.1161/CIRCGENETICS.114.000650 . PMC   4334693 . PMID   25406240.
  26. Priest, James Rush; Gawad, Charles; Kahlig, Kristopher M.; Yu, Joseph K.; O’Hara, Thomas; Boyle, Patrick M.; Rajamani, Sridharan; Clark, Michael J.; Garcia, Sarah T. K.; Ceresnak, Scott; Harris, Jason; Boyle, Sean; Dewey, Frederick E.; Malloy-Walton, Lindsey; Dunn, Kyla; Grove, Megan; Perez, Marco V.; Neff, Norma F.; Chen, Richard; Maeda, Katsuhide; Dubin, Anne; Belardinelli, Luiz; West, John; Antolik, Christian; Macaya, Daniela; Quertermous, Thomas; Trayanova, Natalia A.; Quake, Stephen R.; Ashley, Euan A. (11 October 2016). "Early somatic mosaicism is a rare cause of long-QT syndrome". Proceedings of the National Academy of Sciences. 113 (41): 11555–11560. doi: 10.1073/pnas.1607187113 . PMC   5068256 . PMID   27681629.