Isogenic human disease models

Last updated

Isogenic human disease models are a family of cells that are selected or engineered to accurately model the genetics of a specific patient population, in vitro . They are provided with a genetically matched 'normal cell' to provide an isogenic system to research disease biology and novel therapeutic agents. [1] They can be used to model any disease with a genetic foundation. Cancer is one such disease for which isogenic human disease models have been widely used.

Contents

Historical models

Human isogenic disease models have been likened to 'patients in a test-tube', since they incorporate the latest research into human genetic diseases and do so without the difficulties and limitations involved in using non-human models. [2]

Historically, cells obtained from animals, typically mice, have been used to model cancer-related pathways. However, there are obvious limitations inherent in using animals for modelling genetically determined diseases in humans. Despite a large proportion of genetic conservation between humans and mice, there are significant differences between the biology of mice and humans that are important to cancer research. For example, major differences in telomere regulation enable murine cells to bypass the requirement for telomerase upregulation, which is a rate-limiting step in human cancer formation. As another example, certain ligand-receptor interactions are incompatible between mice and humans. Additionally, experiments have demonstrated important and significant differences in the ability to transform cells, compared with cells of murine origin. For these reasons, it remains essential to develop models of cancer that employ human cells. [3]

Targeting vectors

Isogenic cell lines are created via a process called homologous gene-targeting. Targeting vectors that utilize homologous recombination are the tools or techniques that are used to knock-in or knock-out the desired disease-causing mutation or SNP (single nucleotide polymorphism) to be studied. Although disease mutations can be harvested directly from cancer patients, these cells usually contain many background mutations in addition to the specific mutation of interest, and a matched normal cell line is typically not obtained. Subsequently, targeting vectors are used to 'knock-in' or 'knock out' gene mutations enabling a switch in both directions; from a normal to cancer genotype; or vice versa; in characterized human cancer cell lines such as HCT116 or Nalm6. [4]

There are several gene targeting technologies used to engineer the desired mutation, the most prevalent of which are briefly described, including key advantages and limitations, in the summary table below.

TechniqueGene Knock-InGene Knock-out
rAAV (recombinant adeno-associated virus vectors) [5] Targeted insertions or modifications are created within endogenous genes; and so are subject to:
  1. The correct gene-regulation mechanisms; and
  2. Accurately reflect the disease events found in real patients.

rAAV can introduce subtle point mutations, SNPs as well as small insertions with high efficiency. Moreover, many peer reviewed studies have shown that rAAV does not introduce any confounding off target genomic events.[ citation needed ]

Appears to be the preferred method being adopted in academia, Biotech and Pharma on a precision versus time versus cost basis.[ citation needed ]|

Gene knockouts are at the endogenous locus, and thus are definitive, stable and patient relevant. No confounding off-target effects are elicited at other genomic loci. It requires a 2- step process:
  1. Generate a heterozygous KO
  2. Generate a bi-allelic knockout by targeting the second allele.

This process can therefore generate 3 genotypes (+/+; -/+ and -/-); enabling therefore the analysis of haplo-insufficient gene function.

Current limitation is the need to sequentially target single alleles making generation of knock-out cell lines a two-step process.|

Plasmid-based homologous recombinationInsertion is at the endogenous locus and has all the above benefits, but it is very inefficient. It also requires a promoterless drug selection strategy entailing bespoke construct generation. A large historical bank of cell lines has been generated using this method which has been displaced by other methods since the mid-1990s.Deletion is at endogenous locus and has all the above benefits, but it is inefficient. It also requires a promoterless drug selection strategy that entails bespoke construct generation
Flip-inThis is an efficient technique that allows the directed insertion of 'ectopic' transgenes at a single pre-defined genomic locus (integration via a FLP recombinase site). This is not a technique for modifying an endogenous locus. Transgenes will usually be under the control of an exogenous promoter, or a partially defined promoter-unit in the incorrect genomic location. Their expression will therefore not be under the same genomic and epigenetic regulation as the endogenous loci, which limits the utility of these systems for studying gene-function. They are however, good for eliciting rapid and stable exogenous gene expression.Not applicable
Zinc-Finger Nucleases (ZFNs)ZFNs have been reported to achieve high rates of genetic knock-outs within a target endogenous gene. If ZFNs are co-delivered with a transgene construct homologous to the target gene, genetic knock-in's or insertions can also be achieved. [6] One potential drawback is that any off-target double strand breaks could lead to random off-target gene insertions, deletions and wider genomic instability; confounding the resulting genotype. [7] However, no measurable increase in the rate of random plasmid integration was observed in human cells efficiently edited with ZFNs that target a composite 24 bp recognition site [6] ZFNs are sequence-directed endonucleases which enable the rapid and highly efficient (up to 90% in a bulk cell population) disruption of both alleles of a target gene, although user- defined or patient relevant loss of-function alterations have not been reported at similar frequencies. Off target deletions or insertions elsewhere in the genome are a significant concern. The speed advantage of obtaining a biallelic KO in one step is also partially mitigated if one still needs to derive a clonal cell line to study gene function in a homogenous cell-population.
MeganucleasesMeganucleases are operationally analogous to ZFN's. There are limitations inherent in their use such as the meganuclease vector design which can take up to 9 months and cost tens of thousands of dollars.[ citation needed ] This makes meganucleases more attractive in high-value applications such as gene therapy, agrobiotechnology and engineering of bioproducer lines.

Homologous recombination in cancer cell disease models

Homologous recombination (HR) is a kind of genetic recombination in which genetic sequences are exchanged between two similar segments of DNA. HR plays a major role in eukaryotic cell division, promoting genetic diversity through the exchange between corresponding segments of DNA to create new, and potentially beneficial combinations of genes.[ citation needed ]

HR performs a second vital role in DNA repair, enabling the repair of double-strand breaks in DNA which is a common occurrence during a cell's lifecycle. It is this process which is artificially triggered by the above technologies and bootstrapped in order to engender 'knock-ins' or 'knockouts' in specific genes 5, 7.

A recent key advance was discovered using AAV-homologous recombination vectors, which increases the low natural rates of HR in differentiated human cells when combined with gene-targeting vectors-sequences.[ citation needed ]

Commercialization

Factors leading to the recent commercialization of isogenic human cancer cell disease models for the pharmaceutical industry and research laboratories are twofold.[ citation needed ]

Firstly, successful patenting of enhanced targeting vector technology has provided a basis for commercialization of the cell-models which eventuate from the application of these technologies.[ citation needed ]

Secondly, the trend of relatively low success rates in pharmaceutical RnD and the enormous costs have created a real need for new research tools that illicit how patient sub-groups will respond positively or be resistant to targeted cancer therapeutics based upon their individual genetic profile.[ citation needed ]

See also

Related Research Articles

Gene knockouts are a widely used genetic engineering technique that involves the targeted removal or inactivation of a specific gene within an organism's genome. This can be done through a variety of methods, including homologous recombination, CRISPR-Cas9, and TALENs.

<span class="mw-page-title-main">DNA repair</span> Cellular mechanism

DNA repair is a collection of processes by which a cell identifies and corrects damage to the DNA molecules that encodes its genome. In human cells, both normal metabolic activities and environmental factors such as radiation can cause DNA damage, resulting in tens of thousands of individual molecular lesions per cell per day. Many of these lesions cause structural damage to the DNA molecule and can alter or eliminate the cell's ability to transcribe the gene that the affected DNA encodes. Other lesions induce potentially harmful mutations in the cell's genome, which affect the survival of its daughter cells after it undergoes mitosis. As a consequence, the DNA repair process is constantly active as it responds to damage in the DNA structure. When normal repair processes fail, and when cellular apoptosis does not occur, irreparable DNA damage may occur, including double-strand breaks and DNA crosslinkages. This can eventually lead to malignant tumors, or cancer as per the two-hit hypothesis.

<span class="mw-page-title-main">Homologous recombination</span> Genetic recombination between identical or highly similar strands of genetic material

Homologous recombination is a type of genetic recombination in which genetic information is exchanged between two similar or identical molecules of double-stranded or single-stranded nucleic acids.

<span class="mw-page-title-main">Bert Vogelstein</span> American oncologist (born 1949)

Bert Vogelstein is director of the Ludwig Center, Clayton Professor of Oncology and Pathology and a Howard Hughes Medical Institute investigator at The Johns Hopkins Medical School and Sidney Kimmel Comprehensive Cancer Center. A pioneer in the field of cancer genomics, his studies on colorectal cancers revealed that they result from the sequential accumulation of mutations in oncogenes and tumor suppressor genes. These studies now form the paradigm for modern cancer research and provided the basis for the notion of the somatic evolution of cancer.

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 alleles in an otherwise heterozygous individual. This expression has important implications for the study of tumorigenesis and lethal recessive alleles. 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.

<span class="mw-page-title-main">Oncogenomics</span> Sub-field of genomics

Oncogenomics is a sub-field of genomics that characterizes cancer-associated genes. It focuses on genomic, epigenomic and transcript alterations in cancer.

<span class="mw-page-title-main">MSH2</span> Protein-coding gene in the species Homo sapiens

DNA mismatch repair protein Msh2 also known as MutS homolog 2 or MSH2 is a protein that in humans is encoded by the MSH2 gene, which is located on chromosome 2. MSH2 is a tumor suppressor gene and more specifically a caretaker gene that codes for a DNA mismatch repair (MMR) protein, MSH2, which forms a heterodimer with MSH6 to make the human MutSα mismatch repair complex. It also dimerizes with MSH3 to form the MutSβ DNA repair complex. MSH2 is involved in many different forms of DNA repair, including transcription-coupled repair, homologous recombination, and base excision repair.

<span class="mw-page-title-main">Gene targeting</span> Genetic technique that uses homologous recombination to change an endogenous gene

Gene targeting is a biotechnological tool used to change the DNA sequence of an organism. It is based on the natural DNA-repair mechanism of Homology Directed Repair (HDR), including Homologous Recombination. Gene targeting can be used to make a range of sizes of DNA edits, from larger DNA edits such as inserting entire new genes into an organism, through to much smaller changes to the existing DNA such as a single base-pair change. Gene targeting relies on the presence of a repair template to introduce the user-defined edits to the DNA. The user will design the repair template to contain the desired edit, flanked by DNA sequence corresponding (homologous) to the region of DNA that the user wants to edit; hence the edit is targeted to a particular genomic region. In this way Gene Targeting is distinct from natural homology-directed repair, during which the ‘natural’ DNA repair template of the sister chromatid is used to repair broken DNA. The alteration of DNA sequence in an organism can be useful in both a research context – for example to understand the biological role of a gene – and in biotechnology, for example to alter the traits of an organism.

<span class="mw-page-title-main">ERCC1</span> Protein-coding gene in the species Homo sapiens

DNA excision repair protein ERCC-1 is a protein that in humans is encoded by the ERCC1 gene. Together with ERCC4, ERCC1 forms the ERCC1-XPF enzyme complex that participates in DNA repair and DNA recombination.

<span class="mw-page-title-main">BCL3</span> Protein-coding gene in the species Homo sapiens

B-cell lymphoma 3-encoded protein is a protein that in humans is encoded by the BCL3 gene.

<span class="mw-page-title-main">Exonuclease 1</span> Protein-coding gene in the species Homo sapiens

Exonuclease 1 is an enzyme that in humans is encoded by the EXO1 gene.

Mouse models of colorectal cancer and intestinal cancer are experimental systems in which mice are genetically manipulated, fed a modified diet, or challenged with chemicals to develop malignancies in the gastrointestinal tract. These models enable researchers to study the onset, progression of the disease, and understand in depth the molecular events that contribute to the development and spread of colorectal cancer. They also provide a valuable biological system, to simulate human physiological conditions, suitable for testing therapeutics.

Synthetic lethality is defined as a type of genetic interaction where the combination of two genetic events results in cell death or death of an organism. Although the foregoing explanation is wider than this, it is common when referring to synthetic lethality to mean the situation arising by virtue of a combination of deficiencies of two or more genes leading to cell death, whereas a deficiency of only one of these genes does not. In a synthetic lethal genetic screen, it is necessary to begin with a mutation that does not result in cell death, although the effect of that mutation could result in a differing phenotype, and then systematically test other mutations at additional loci to determine which, in combination with the first mutation, causes cell death arising by way of deficiency or abolition of expression.

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

Fanconi anemia, complementation group M, also known as FANCM is a human gene. It is an emerging target in cancer therapy, in particular cancers with specific genetic deficiencies.

The NSG mouse is a brand of immunodeficient laboratory mice, developed and marketed by Jackson Laboratory, which carries the strain NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ. NSG branded mice are among the most immunodeficient described to date. NSG branded mice lack mature T cells, B cells, and natural killer (NK) cells. NSG branded mice are also deficient in multiple cytokine signaling pathways, and they have many defects in innate immunity. The compound immunodeficiencies in NSG branded mice permit the engraftment of a wide range of primary human cells, and enable sophisticated modeling of many areas of human biology and disease. NSG branded mice were developed in the laboratory of Dr. Leonard Shultz at Jackson Laboratory, which owns the NSG trade mark.

<span class="mw-page-title-main">Chromothripsis</span> Massive chromosomal rearrangement process linked to cancer

Chromothripsis is a mutational process by which up to thousands of clustered chromosomal rearrangements occur in a single event in localised and confined genomic regions in one or a few chromosomes, and is known to be involved in both cancer and congenital diseases. It occurs through one massive genomic rearrangement during a single catastrophic event in the cell's history. It is believed that for the cell to be able to withstand such a destructive event, the occurrence of such an event must be the upper limit of what a cell can tolerate and survive. The chromothripsis phenomenon opposes the conventional theory that cancer is the gradual acquisition of genomic rearrangements and somatic mutations over time.

Recombinant adeno-associated virus (rAAV) based genome engineering is a genome editing platform centered on the use of recombinant AAV vectors that enables insertion, deletion or substitution of DNA sequences into the genomes of live mammalian cells. The technique builds on Mario Capecchi and Oliver Smithies' Nobel Prize–winning discovery that homologous recombination (HR), a natural hi-fidelity DNA repair mechanism, can be harnessed to perform precise genome alterations in mice. rAAV mediated genome-editing improves the efficiency of this technique to permit genome engineering in any pre-established and differentiated human cell line, which, in contrast to mouse ES cells, have low rates of HR.

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.

Mutational signatures are characteristic combinations of mutation types arising from specific mutagenesis processes such as DNA replication infidelity, exogenous and endogenous genotoxin exposures, defective DNA repair pathways, and DNA enzymatic editing.

<span class="mw-page-title-main">Alberto Bardelli</span> Italian geneticist

Alberto Bardelli is an Italian geneticist and cancer researcher, expert in the field of precision medicine. He is a full professor of histology at the Department of Oncology, University of Turin and Scientific Director of IFOM, the AIRC Institute of Molecular Oncology.

References

  1. Torrance CJ, Agrawal V, Vogelstein B, Kinzler KW (October 2001). "Use of isogenic human cancer cells for high-throughput screening and drug discovery". Nat. Biotechnol. 19 (10): 940–5. doi:10.1038/nbt1001-940. PMID   11581659. S2CID   21633547.
  2. Gupta, Piyush B.; Kuperwasser, Charlotte (2004). "Disease models of breast cancer". Drug Discovery Today. 1: 9–16. doi:10.1016/j.ddmod.2004.05.001.
  3. Hirata R, Chamberlain J, Dong R, Russell DW (July 2002). "Targeted transgene insertion into human chromosomes by adeno-associated virus vectors". Nat. Biotechnol. 20 (7): 735–8. doi:10.1038/nbt0702-735. PMID   12089561. S2CID   11572520.
  4. Masters JR (December 2000). "Human cancer cell lines: fact and fantasy". Nat. Rev. Mol. Cell Biol. 1 (3): 233–6. doi:10.1038/35043102. PMID   11252900. S2CID   21839266.
  5. Engelhardt JF (August 2006). "AAV hits the genomic bull's-eye". Nat. Biotechnol. 24 (8): 949–50. doi:10.1038/nbt0806-949. PMID   16900138. S2CID   26734696.
  6. 1 2 Urnov, Fyodor D.; Rebar, Edward J.; Holmes, Michael C.; Zhang, H. Steve; Gregory, Philip D. (2010). "Genome editing with engineered zinc finger nucleases". Nature Reviews Genetics. 11 (9): 636–646. doi:10.1038/nrg2842. PMID   20717154. S2CID   205484701.
  7. Radecke S, Radecke F, Cathomen T, Schwarz K (April 2010). "Zinc-finger nuclease-induced gene repair with oligodeoxynucleotides: wanted and unwanted target locus modifications". Mol. Ther. 18 (4): 743–53. doi:10.1038/mt.2009.304. PMC   2862519 . PMID   20068556.

Sources