Journal Pre-proof Technology used to build and transfer mammalian chromosomes David M. Brown, John I. Glass PII:
S0014-4827(20)30045-8
DOI:
https://doi.org/10.1016/j.yexcr.2020.111851
Reference:
YEXCR 111851
To appear in:
Experimental Cell Research
Received Date: 8 November 2019 Revised Date:
9 January 2020
Accepted Date: 14 January 2020
Please cite this article as: D.M. Brown, J.I. Glass, Technology used to build and transfer mammalian chromosomes, Experimental Cell Research (2020), doi: https://doi.org/10.1016/j.yexcr.2020.111851. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Inc.
David M. Brown: Conceptualization, Writing - Original Draft, Writing - Review & Editing, Visualization. John I. Glass: Conceptualization, Writing - Review & Editing, Funding acquisition.
1
Technology used to build and transfer mammalian chromosomes
2
David M. Brown1 and John I. Glass2*
3 4
1
5
2
J. Craig Venter Institute, Rockville, Maryland, United States of America J. Craig Venter Institute, La Jolla, California, United States of America
6 7
* Corresponding author
8
E-mail:
[email protected]
9 10 11
Keywords
12
Spheroplast fusion, Microcell mediated chromosome transfer (MMCT), mammalian artificial
13
chromosome (MAC), human artificial chromosome (HAC), transformational-associated
14
recombination (TAR) cloning
15 16 17 18 19 20 21 22 23 24 25 26
27
Abstract
28
In the near twenty-year existence of the human and mammalian artificial chromosome field, the
29
technologies for artificial chromosome construction and installation into desired cell types or
30
organisms have evolved with the rest of modern molecular and synthetic biology. Medical,
31
industrial, pharmaceutical, agricultural, and basic research scientists seek the as yet unrealized
32
promise of human and mammalian artificial chromosomes. Existing technologies for both top-
33
down and bottom-up approaches to construct these artificial chromosomes for use in higher
34
eukaryotes are very different but aspire to achieve similar results. New capacity for production
35
of chromosome sized synthetic DNA will likely shift the field towards more bottom-up
36
approaches, but not completely. Similarly, new approaches to install human and mammalian
37
artificial chromosomes in target cells will compete with the microcell mediated cell transfer
38
methods that currently dominate the field.
39 40
Introduction
41
Human and mammalian artificial chromosomes (HACs and MACs respectively) have been
42
important research areas for more than twenty years. The possibility of adding new
43
chromosomes comprising megabases of DNA to cells opens many opportunities. MAC/HACs
44
offers advantages that other approaches to gene therapy and transgenic animal production do
45
not. For instance, unlike approaches using retro and lentiviruses, when using MAC/HACs to
46
genetically alter cells, the native genome of cells is unaltered. Furthermore, the size of
47
MAC/HAC gene payload is much more than the 10 kb limit of most viral based gene therapy
48
approaches, which makes it possible to consider adding hundreds of new genes to cells or to
49
think about including entire corrected versions of damaged genes, such as the 2.4 Mb
50
dystrophin gene responsible for Duchene muscular dystrophy. Unfortunately, the promise of
51
MAC/HACs for gene therapies and other applications remains unfulfilled, even though
52
mammalian and human artificial chromosomes have potential that could have a dramatic effect
53
on healthcare.
54 55
For a MAC/HAC to be useful in gene therapy it needs to be stable, single copy, easy to
56
manipulate, and capable of carrying a payload of genetic material. Herein we summarize and
57
review the technologies to generate and deliver MAC/HACs and how each technique can be
58
utilized in the scheme to develop gene therapies. There have been continued advancements in
59
several areas including DNA transfer and assembly technologies. These technologies can be
60
linked together to create a web of different pathways for moving synthetic DNA or natural
61
chromosomes to different cell types and editing them to be used for various applications. Some
62
techniques are better than others and some need more refining, however manipulation of
63
synthetic chromosomes may soon enter mainstream use.
64 65
The hope for MAC/HACs is that the field will develop as yeast artificial chromosomes (YACs)
66
did in the 1980s (1). The identification of the yeast centromere and autonomous replication
67
sequence led to a revolution of genetic tools and capabilities in yeast. That has had a dramatic
68
effect in the field of synthetic biology and led to a catalytic expansion in our understanding of
69
yeast biology, as well as novel forms of protein expression. Building on the success of YACs,
70
yeast now has advanced genomic tools that can be utilized in the construction of other synthetic
71
chromosomes. Particularly, yeast can be used to generate HACs using a bottom-up technique
72
that uses synthetic DNA to assemble a HAC. First, synthetic DNA can be constructed in vitro
73
and then either transfected into a mammalian cell culture or transformed into yeast to produce
74
larger, more refined constructs. The DNA can then be transferred to a mammalian cell via yeast
75
fusion or by collecting the DNA from yeast and transfecting to the mammalian cells. Once
76
present in a recipient cell, the HAC containing cell can be transferred or reprogramed to the cell
77
type needed. The aforementioned bottom-up techniques describe one of two general
78
approaches to HAC generation. The second being “top-down:” in this approach, natural HACs
79
are whittled down to the functional components and then payload DNA is added to the HAC.
80
This technique mainly relies on microcell mediated chromosome transfer (MMCT) to first edit
81
natural chromosomes in capable cell lines and then transfer to the cell line of choice.
82 83
In vitro DNA synthesis and engineering
84
The emerging field of synthetic biology has resulted from a dramatic and rapid drop in the costs
85
of DNA synthesis, which has reinvigorated the effort to generate useful HACs. Researchers can
86
now order small custom genes for a few hundred dollars and receive them in just a few weeks
87
(2, 3). The synthesis of DNA has become a commodity with numerous vendors capable of
88
producing highly complex sequences that were impossible to synthesize just 15 years ago.
89
Construction of megabase sized chromosomes has been demonstrated (4, 5).
90
Robust methods of DNA synthesis and assembly can now be utilized in HAC assembly or as
91
part of HAC editing. The development of approaches for synthesis of longer oligonucleotides
92
containing fewer errors has resulted in DNA foundries synthesizing larger and larger pieces of
93
DNA for rapid use. Contracting DNA foundries to synthesize DNA payloads is not the only
94
option. Competent molecular biology labs can readily synthesize 1-3 kb DNA segments from
95
oligos (6), and then assemble those segments into larger molecules. Many methods to
96
assemble oligos or larger DNA fragments rely on “homology-based assembly.” One method
97
referred to as Gibson Assembly (4-7) uses the T5 exonuclease to chew-back so that
98
homologous ends can then anneal to the overlap regions. Other enzymes then join and repair
99
the fragments.
Numerous other homology based methods have also been developed (8),
100
including overlapping PCR (9), CPEC (10), SLIC (11), SLiCE (12),SIRA (13), USER (14), as
101
well as many commercial kits available, including In-Fusion (Clontech), Gateway and GeneArt
102
(Life Technologies), and NEBuilder® HiFi DNA Assembly (NEB).
103
assembly based on Restriction digestion/ligation assembly (15) such as Biobrick (16, 17),
104
golden gate (18) and LCR (19) are available. However, the trend towards cheaper commercial
105
DNA synthesis means any group thinking of synthesizing HAC payloads must weigh the relative
106
material and labor cost of doing it in house or contracting a foundry to make the needed DNA.
Other methods of DNA
107 108
Yeast assembly
109
Brewer’s yeast, Saccharomyces cerevisiae, is an organism with extensive genetic tools thanks
110
to its recombination capacity (1, 20) and, as already discussed, modern yeast genetics enable
111
manipulation and construction of large DNA molecules as linear YACs or as circular yeast
112
centromeric plasmids (YCPs). This means that yeast is an excellent organism in which to
113
construct and then to deliver those molecules into mammalian cells. The primary process of
114
manipulating synthetic DNA in yeast is transformational-associated recombination (TAR) cloning
115
(1).
116
assemblies including HACs (5, 7, 20-23), and synthetic yeast chromosomes (24-33).
117
technique works by utilizing the recombination capacity of yeast to assemble overlapping DNA
118
molecules (Figure 1).
119
Originally TAR cloning was developed for genomic studies (1), where yeast could recover
120
chromosome segments by “fishing out” segments of genomic DNA as large as 300 kb, which
121
could then be analyzed further and create libraries of different genes. However TAR cloning is
122
now frequently used in synthetic biology applications, including producing synthetic genomes,
TAR cloning is a powerful technique that can rapidly produce large synthetic DNA The
123
and can be modified for more specialized assemblies (34, 35). The design of each of the TAR
124
cloning fragments would be the same as for the in vitro homology-based methods described
125
above, such as Gibson Assembly.
126
homologous ends to each adjacent segment are constructed.
127
recombine these segments together to form larger molecules. The flexibility and robustness of
128
TAR cloning has been repeatedly demonstrated. Gibson and colleagues transformed yeast with
129
25 overlapping fragments between 17 and 35 kb in length (7).
130
have been assembled (20, 36), and oligonucleotide assembly with as many as 38 overlapping
131
oligonucleotides with only 20 base overlaps into a plasmid has also been reported (37).
132
Another advantage with TAR cloning compared to in vitro methods is a selected gene can be
133
used to “fish out” a gene of interest from genomic DNA (1), meaning the homologous sections of
134
DNA do not need to be strictly at the 5’ or 3’ ends but will recombine and assemble across the
135
homologous regions to form an assembly, although with a considerable drop in efficiency.
Synthetic DNA or PCR amplified DNA containing Yeast spontaneously will
High G/C content DNA YACs
136 137
Additionally, TAR cloning can be used to build de novo HACs produced in yeast using a bottom-
138
up strategy (38). Ebersole et al. transfected yeast synthetic human alpha satellite (alphoid)
139
DNA tandem repeats to produce long tandem arrays of up to 120 kb that in mammalian cells,
140
associated with centromere proteins like CENP-A, form competent HACs (39). This technique
141
has been used to generate a HAC with a conditional centromere, (40, 41) and is described in
142
this review later as the alphoidtetO-HAC. In one example of combining the capabilities of TAR
143
cloning, a portable BRCA1-HAC module for analysis of BRCA1 tumor suppressor function has
144
been produced (42). Production of the BRCA1-HAC first involved the recovery of the BRCA1
145
gene into a YAC and then insertion into a HAC.
146 147
148
Construction of mammalian chromosomes
149
Knowledge about centromere structure, formation and stability is essential to design and build
150
effective HACs. The understanding of centromere formation and de novo HAC formation has
151
leapt forward in recent years. The structural components that assemble a centromere on
152
chromatin are largely understood; however, the centromere organization and dynamics still
153
need study. Excellent reviews on the subject have been published previously (43-48), but
154
briefly, some of the most significant findings include: the discovery of a 171 bp repetitive
155
sequence around where the centromere forms was identified and termed alpha satellite
156
(alphoid) DNA (49). Identification of CENP-A, a H3 histone, that forms a centromere specific
157
nucleosome with H2A, H2B and H4 and is actively replenished to maintain centromere identity
158
around alphoid DNA (45), and is what defines a functional HAC (51, 52) was a critical discovery.
159
CENP-B was the next major protein to be identified, and is the only protein that actually binds
160
alphoid DNA, specifically, to a 17bp motif termed the CENP-B box (53). In total over 100
161
centromere/kinetochore protein components have been identified. Another important protein is
162
HJURP, which recruits CENP-A to the centromere and is stabilized by a number of remodeling
163
factors (54-57). Several models for maintenance and organization of the centromere also exist
164
(43, 58, 59).
165 166
As stated previously, there are two general methods of constructing HACs: “top-down” and
167
“bottom-up”. Top-down HACs focus on engineering HACs from existing chromosome structures
168
and has been reviewed previously (44, 60-63). Small chromosomes such as the human Y
169
chromosome are whittled down to a more manageable size for experimentation.
170
editing/winnowing down is typically performed based on telomere-directed chromosome
171
truncation in the homologous recombination proficient chicken DT40 cell line (64). Editing is
172
possible because of the notable advancement of inserting a loxP landing pad site into a HAC
The
173
present in DT40 cells, allowing the HAC to be manipulated using Cre-loxP recombination (41,
174
65). Minichromosomes have been produced from the human chromosomes X, Y, 14 and 21
175
(64, 66-68).
176 177
De novo HACs, or “bottom-up” engineered HACs, use alphoid DNA cloned in bacterial or yeast
178
artificial chromosomes that are transfected into mammalian cells, which multimerize and form
179
stable HACs 1-10 Mb as a result of the amplification of alphoid DNA. First generation HACs,
180
such as in Harrington et al. (69), was produced from arrays of a 2.7 kb repeats from
181
chromosome 17 to produce a final linear 173 kb product. Upon transfection into HT1080 cells,
182
this HAC further multimerized and formed HACs 6-10 Mb. Once expanded to that size, the
183
HAC stably replicated and segregated for over 6 months as determined by FISH. The principle
184
of using repeated segments of alphoid DNA was deployed in other first generation HACs (70),
185
and has generally remained today. The first observation that alphoid repeats are sufficient for
186
CENP-A assembly in many cell types, enabled the first de novo HAC construction. However, a
187
recent study by Black and colleagues demonstrated that a non-repetitive sequence (4q21) has
188
been shown to form functional HACs without CENP-B boxes or alphoid DNA at low rates (71).
189
Meaning the flexibility of how centromeres can form may be greater than previously thought.
190
The rate of centromere formation on 4q21 can be increased and become possible on alphoid
191
DNA is by a single pulse of epigenetic seeding of centromeric chromatin. In this case LacI-
192
HJURP targeted to arrays of LacO repeats, leading to formation of CENP-A nucleosomes (71).
193
Thus, it appears there are at least three ways to form a competent mammalian centromere.
194
First, alphoid DNA alone can form HACs (52, 69). Second, seeding of CENP-A nucleosomes
195
can form HACs (71) and third, a non- alphoid DNA (4q21) can lead to HAC formation (71).
196
197
Another notable advancement in HAC technology is the development of the alphoidtetO-HAC.
198
(72) In this HAC, a ~50 kb synthetic alphoid DNA array and a tetracycline operator (tetO) were
199
combined to form a conditional centromere (40, 41, 73) such that when a fusion protein of the
200
Tet Repressor (TetR) with a KRAB silencing domain (tTS) (74) is expressed the kinetochore
201
disassociates and the HAC can be lost. This alphoidtetO-HAC was used in subsequent studies
202
to determine how kinetochore formation occurs and what other proteins are involved in the
203
process (75).
204 205
Until recently, de novo HAC formation has been limited to very few human cell lines, notably
206
HT1080 (76, 77). However, Ohzeki et al. (52) discovered that the H3K9 acetyl/methyl balance
207
explains the cell specificity for de novo HAC formation. Specifically, that H3 methylation (H3K9
208
methylation) is necessary for HP1 assembly, which is necessary for heterochromatin formation,
209
and that the ratio of H3k9me3 is different dependent on cell type, with HeLa cells having a high
210
ratio and HT1080 cells having a low ratio.
211
centromere can significantly stabilize the HAC, thus allowing for the expansion of de novo HAC
212
formation to other cell types (52).
Tethering histone acetyl transferase to the
213 214
Delivery of synthetic DNA to mammalian cells by yeast fusion
215 216
As construction of these bottom-up MAC/HACs and large DNA constructs becomes easier and
217
more routine to make, the delivery of large segments of DNA to the mammalian nucleus
218
remains a limiting step to effective gene therapy.
219
cells, yeast spheroplasts (yeast whose cell walls were enzymatically removed) can be fused by
220
a polyethylene glycol (PEG) mediated cellular fusion method (78). The advantage of this
To deliver DNA from yeast to mammalian
221
method is that yeast can be used as a construction platform to build large DNA molecules (4,
222
79, 80) that can then be delivered to mammalian cells. The technique was first developed in the
223
1990s and has since been used to transfer yeast artificial chromosomes to several cell types
224
including goat fibroblasts (81), mouse A9 cells (82), mouse ES cells (83, 84), human renal
225
carcinoma cells (85), and human epithelial HEK293 cells (86, 87).
226
excess of 500 kb has been used to create transgenic mice through spheroplast fusion with
227
mouse embryonic stem cells (82).
228
technique (83, 85, 88-90). However, previously we developed an improved method of fusing
229
yeast spheroplasts by arresting the recipient mammalian cell in the mitotic phase (M-phase) of
230
the cell cycle when the mammalian nuclear envelope is broken down, thus allowing for easier
231
diffusion of the vector to where the mammalian nucleus re-forms (78). Importantly, the vector
232
as yeast chromatin seems to be largely recognized by mammalian RNA polymerase and
233
expressed, as evidenced by the recognition of delivered herpes simples virus (HSV-1) genomes
234
and production of HSV-1 virions via yeast fusion (78). Interestingly, DNA delivery rates of
235
vector sizes up to 1.1 Mb do not impede the DNA delivery rate and were delivered at similar
236
rates to smaller vectors. The primary advantage of this method over lipofection and
237
microinjection is its capacity to deliver larger DNA constructs.
238
molecules to shear forces leads to breakage, decreasing delivery efficiency. This means that
239
the use of agarose plugs is necessary to avoid DNA damage due to shear (91-93). In contrast,
240
PEG-mediated fusion does not require DNA isolation and thus eliminates exposure of DNA to
241
shear damage and potential breakage. Recently the 16 chromosomes of Saccharomyces
242
cerevisiae were stitched together to form a single linear chromosome 11.8 Mb in size, which is
243
the largest reported yeast chromosome to date (94), and the strain did not suffer significant
244
growth problems. Another group also found a similar result reducing the number of yeast
245
chromosomes to 2, each approximately 6 Mb (30). These experiments suggest it may be
246
possible to construct HACs as YACs comprising millions or even tens of millions of basepairs.
In addition, yeast DNA in
This is conventionally a very low efficiency delivery
Exposure of large DNA
247 248
Generally, the mechanism by which two membranes fuse involves two steps: aggregation and
249
membrane merging. Two membranes will naturally aggregate via Van der Waals attraction
250
without the hydration electrostatic forces created by water molecules that creates a strong
251
repulsive barrier keeping the phospholipid membrane intact.
252
binding sites or lowering the number of charges on the membrane leads to aggregation of
253
nearby membranes (95, 96). Once membranes aggregate, the fusion mechanism still remains
254
partially unknown, but one hypothesis is that the membranes will fuse when at least two regions
255
with transient and sufficient bilayer disorder temporarily come into close contact (95). Meaning
256
that regions of temporary membrane compositional disorder are required for successful cell
257
fusion, and that membrane proteins can provide these defects. Without these defects, the outer
258
membrane bilayer leaflets will merge, while the inner leaflets remain distinct and no mixing of
259
the aqueous layer is observed (97).
Lowering the number of water
260 261
PEG mediated cell fusion allows for easier installation of yeast borne HACs into mammalian
262
cells: however, calcium, diacylglycerol, peptides, or high membrane curvature, can also induce
263
fusion (97, 98). Saez et al. and Boni et al. showed by electron microscopy and X-ray scattering
264
respectively that PEG-based fusion primarily functions by volume exclusion, resulting in an
265
osmotic force that aggregates membranes together in a dehydrated region (98, 99). PEG-
266
mediated cell fusion has been widely adopted for a wide range of cell types, although
267
optimization is often needed for effective delivery and to avoid toxicity and cell lysis. The
268
process has been shown to result in transfer of DNA to and from bacterial protoplasts to
269
bacterial protoplasts (5, 100-104), from bacterial mini cells to yeast spheroplasts (105, 106),
270
from bacterial protoplasts to mammalian cells (93, 107, 108), from yeast spheroplasts to
271
mammalian cells (78, 82, 87, 109, 110), from yeast spheroplasts to insect cells (78), and from
272
yeast spheroplasts to avian cells (78).
Additionally, very large DNA molecules have been
273
transferred this way. A 1.1 Mb bacterial genome cloned in yeast was transplanted from into a
274
different species bacterial cell (79, 104), and vectors in excess of 1 Mb have been transferred
275
into mammalian cells from yeast spheroplasts (78, 82).
276 277
The implications of this technique are that bottom-up HACs can be first constructed in yeast and
278
then delivered to the mammalian cell without having to harvest in vitro DNA, minimizing shear
279
forces and enabling the construction of larger synthetic DNA, over 1 Mb and perhaps
280
significantly over 1 Mb. However, it is likely that top-down HACs are not amenable to yeast
281
fusion, as the vectors are already present in mammalian cell lines, and these HACs are larger
282
than the largest reported YACs (89).
283 284
Microcell mediated chromosome transfer
285
Microcell mediated chromosome transfer (MMCT) is a technique by which entire chromosomes
286
are transferred from one mammalian cell to another by cellular fusion. It has been critical for
287
development of HACs using top-down approaches. The technique was first developed in 1977
288
(111). Traditionally, the process begins with arresting donor cells in metaphase using colcemid
289
or colchicine as a microtubule elongation inhibitor and depolymerizer. During a transition period
290
certain cells transition from metaphase to a pseudo G1 phase, called micronuclei (112, 113), a
291
process by which a membrane forms around individual chromosomes. The process requires a
292
donor cell that can form microcells, which is generally very limited. The most commonly used
293
cell lines are Chinese hamster ovary (CHO) cells, A9 cells and DT40 cells. Most other cell lines
294
die under prolonged exposure to microtubule inhibitors. Actin microfilaments are then disrupted
295
with cytochalasin B and microcells are centrifuged through a Percoll gradient, which prevents
296
cells from entering interphase (112). The purified microcells are then filtered through a 3 µm
297
filter to select for microcells containing just one chromosome. Microcells can then be fused to
298
the recipient cell through PEG-mediated fusion. After selection, desired cell hybrids can be
299
identified. However, this traditional method suffered from very low efficiency (83, 85, 90).
300
alternative to the extended use of microtubule inhibitors was originally developed in 1975, by
301
placing mitotic cells at 4°C followed by resuming g rowth at 37°C. An expanded number of cell
302
lines, including HeLa cells, would create “mini-segrants” that included abnormal chromosome
303
segregations very similar to microcells (114).
304
pursued once MMCT was developed. Recently, several higher efficiency MMCT approaches
305
have been developed including: 1) replacement of colcemid (microtubule inhibitor) and
306
cytochalasin B (actin destabilizer) with alternates like TN-16 + Griseofluvin and Latrunculin B,
307
which resulted in a 6x higher efficiency (115); 2) using CHO cells to express envelope proteins
308
derived from ecotropic or amphotropic murine leukemia viruses, which resulted in a 26x
309
increase in MMCT transfer rate (116); and finally 3) by retargeting the microcell fusion by adding
310
anti-Transferrin receptor (TfR) single chain antibodies (scFvs) to the extracellular C-terminus of
311
the measles virus hemagglutinin protein, which has been reported to improve the efficiency of
312
MMCT to human fibroblasts (117).
313
Because the most amenable donor cell lines to MMCT are murine models, transfer of
314
chromosomes to mouse cell lines has been relatively successful.
315
include the creation of A9 or CHO-microcell hybrid libraries that contain individual human
316
chromosomes along with each mouse chromosome; meaning these chimeric mouse cell
317
libraries can provide a resource for the functional expression and mapping of human genes
318
(113, 115, 118, 119). Another major advancement was the transfer of a MAC to mouse
319
embryonic stem cells which generated a transchromosomic mouse as a stable animal model
320
(120).
One
However, this technique was generally not
Notable advancements
These can have numerous applications such as the generation of knock out or
321
transchromosomic murine models and humanized animal models (121).
Notably, the capacity
322
of yeast fusion to deliver synthetic DNA to CHO cells (78) means HACs cloned as YACs may be
323
“mobile” within MMCT, possibly linking the two techniques.
324 325
Other methods of chromosome transfer
326
Other methods of chromosome transfer exist; however, these are only possible using highly
327
specialized technicians and equipment. In addition, chromosomes to be transferred must be
328
prepared very gently. Normally the cells in which the chromosomes are constructed are
329
suspended in agarose plugs, then the chromosomes are freed from their cells by proteinase and
330
detergent treatment so that the desired large DNA molecules are safe from fluid shear forces in
331
tiny agarose caverns. To use the chromosomes, the agarose plugs are gently melted or
332
dissolved and the DNA is only pipetted with extreme gentleness (91-93). The most notable of
333
these other methods is needle microinjection. In this process, there is direct injection of DNA
334
into the nucleus of cells. This technique utilizes a thin glass microneedle to puncture both the
335
cell membrane as well as the nuclear envelope.
336
enough for the DNA vector to diffuse across the length of the needle into the nucleus.
337
Microinjection has been common practice since the 1980s (122). The technique has been used
338
to produce recombinant cell lines (123), transgenic animals (124), and for mitochondrial DNA
339
transfer (125).
The needle remains inside the cell long
340 341
Modifications of chromosomes
342
Once chromosomes are present in yeast they can be modified easily. Cre-LoxP technology is a
343
widely used site specific DNA recombination system originally derived from bacteriophage P1
344
(126, 127).
Cre recombinase catalyzes a recombination reaction between two loxP sites
345
requiring no other accessory factors. The loxP site comprises two 13 base inverted repeats
346
separated by an 8 base asymmetric spacer sequence for a total of 34 bases (126, 128). This
347
Cre-LoxP system has been utilized in yeast (129) and more recently has been used in the
348
construction of synthetic yeast chromosomes in the synthetic Sc2.0 project with all non-
349
essential genes flanked by loxPsym sites. Cre recombinase is then induced and rearranges the
350
genome, referred to as SCRaMbLE (25, 27), allowing for the study of genome organization
351
yeast evolution etc. This systematic process of using an enzyme to generate a site-specific
352
clean exchange of a cassette for an analogous cassette of choice is termed recombinase
353
mediated cassette exchange (RMCE). RMCE was originally based on the yeast integrase Flp
354
but has now mostly been replaced by the more active Cre recombinase (130). Since the
355
development of RMCE, there have been improvements such as a dual RMCE (130-132) and
356
cell lines that improve the efficiency of integration (133).
357
methods rely on the principles of TAR cloning include TREC (34), TREC-IN (35), and the
358
CRISPR based methods (134, 135) to edit, add and remove segments of DNA. Recent progress
359
using genome engineering in using CRISPR-Cas systems has also been made (136-140). All
360
these editing methods primarily rely on using the efficient double strand break and repair system
361
contained within yeast (141-144).
Other yeast based DNA modification
362 363
Although yeast has inherent and powerful modification capacity, modifications of mammalian
364
chromosomes in cell culture is now possible using Cre-loxP as well (64, 65, 145). The chicken
365
cell line DT40, a recombination competent cell line, has primarily been the main cell line used to
366
edit chromosomes using loxP technology (65, 66). In those studies, top-down HACs were
367
originally derived from natural chromosomes and edited in a top-down manner. These
368
chromosomes are then “mobile” because DT40 cells can be used for MMCT, so chromosomes
369
can be transferred to and from CHO cells and murine A9 cells. These edited chromosomes can
370
also be transferred into many recipient cell lines including murine ES cells which could be used
371
to generate HAC containing mice (41, 65, 116, 145-148).
372 373
Editing using CRISPR has been reported on plasmids using the human cell line HEK 293 cells
374
(149-151) and has continued to develop rapidly (152) with new and advanced methods being
375
reported constantly. Some important developments include CRISPR/Cas9-mediated genome
376
editing in nonhuman primates (153), modifications to MAC/HACs (154) within the cell lines CHO
377
and A9, connecting the technique to cell lines that are MMCT capable (147). Others have
378
reviewed other less popular editing methods such as zinc finger nucleases (ZFN), and
379
transcription activator-like effector nucleases (TALEN) for genome engineering (155) that have
380
also been applied to mammalian cell culture, and production of transgenic animals (156, 157).
381 382 383
Applications and Differentiated cells
384
As noted above, one of the greatest hopes for HACs is gene therapy applications. This could
385
include using cell-based therapies or perhaps to harvest stem cells from patients and install in
386
them a set of great single chain antibodies to protect them against new infectious diseases or
387
common cancers. Applications are limited only by imagination and limitations in HAC
388
technology. The transfer of artificial chromosomes to useful recipient cell lines is essential for
389
robust gene therapy applications. Several techniques exist; however, they are not robust for all
390
possible applications. Transfer of HACs to induced pluripotent stem cells (158-161) would be
391
very useful for gene therapies because these cells can first be produced from a patients
392
fibroblasts and then differentiated to the cell type of choice. A direct transfer of chromosomes
393
using MMCT to iPSC (158, 160, 162, 163) is also being attempted. Recently, Sinenko et al.
394
reported that the transfer of HACs to iPSC cells could be done by retro-MMCT and maintained
395
as an autonomous chromosome without affecting the pluripotent properties of the cells (162).
396
Although when speed is necessary, direct reprograming of differentiated somatic cells might be
397
of more use, because it reduces the time required by combining two reprograming steps to one.
398
For example, using lineage-specific factors to directly convert fibroblasts into neuronal cells
399
(164).
400 401
Somatic nuclear transfer to embryos (63, 165-168) can be used to clone transgenic animals or
402
to differentiate the embryos to the target cell line. This might be a useful technique to transfer a
403
MAC/HAC to a pluripotent cell type or embryo to create chimeric animals. In this process the
404
nucleus of a donor mammalian cell is removed and then implanted in enucleated oocytes of the
405
target mammal. It has primarily been used in cloning mammals (165, 167, 169). However in
406
some cases it has occasionally been used to for germline modification (170) or for transfer of
407
artificial chromosomes (171). However it is generally a difficult technique with low efficiency, 5-
408
15% (166).
409 410
Applications that involve the delivery of DNA along with functional protein could be developed
411
using the yeast fusion DNA delivery method. The delivery technology has already been used to
412
deliver active fluorescent protein with the DNA vector as a proof of concept. Additionally, when
413
the HSV-1 genome was delivered along with Ebola VP35 to the same mammalian cell there
414
was a differential in infectivity between different cell lines for HSV-1 generation (78). A yeast
415
strain engineered to contain the entire HSV-1 genome and to also express the Ebola derived
416
VP35, which blocks the host cell antiviral response (172, 173), could shut down host cell
417
interferon response and obtain viral replication in cells which HSV-1 had not replicated. Yeast
418
fused with Vero cells and HeLa cells showed differentials in HSV-1 generation levels depending
419
on whether VP35 was included or not (78).
420 421
Conclusion
422
Since
423
microchromosomes in the late 1990s (69), the field has made huge progress. Today, basic
424
MAC/HAC tools are in place and multiple methods can be utilized to construct and deliver
425
HACs, although there is need for much improvement. Figure 3 depicts a schematic of the
426
technologies summarized in this article and associated with the two basic approaches to
427
MAC/HAC construction and installation, “top-down” and “bottom-up.” Huge advances in DNA
428
synthesis have already made construction of megabase sized DNA molecules affordable to
429
many labs and eventually may make such synthetic molecules the only way to produce artificial
430
chromosomes. These advances are moving mainstream MAC/HAC research towards bottom-up
431
approaches, but for the foreseeable future, both top-down, bottom-up and hybrid approaches
432
will be in use.
MAC/HAC
research
dawned
with
the
first
generation
of
human
artificial
433 434
Where will MAC/HAC science be going? For some years now, some synthetic biologists have
435
advocated for a project to synthesize chromosomes, even genomes of a higher eukaryote,
436
possibly even a human genome (174). As noted above, DNA synthesis technology is steadily
437
improving. The Engineering Biology Research Consortium document, “A Research Roadmap
438
for the Next-Generation BioEconomy” projects huge advances in the size, the per nucleotide
439
error rates of synthetic oligonucleotides, and dramatic improvements in single and multiple
440
fragment DNA assembly such that whole chromosomes and genomes can be readily
441
produced within 10 years (175).
As predicted, advances in DNA synthesis will lead to
442
advances in MAC/HAC synthesis and likely make bottom-up chromosome construction the
443
norm. The race to rapidly and cheaply producing large synthetic DNA molecules has many
444
academic and industrial competitors, theseemerging technologies with real promise, are only
445
half of the technical advances needed to bring in an era of efficient gene therapy and
446
production of new drugs and animals using MAC/HACs. Synthetic chromosomes must be
447
installed and booted up in target cells more efficiently than can be done at present before
448
their promise can be realized. However, as we have tried to present in this review, HAC
449
installation technology lags behind DNA synthesis technology. More research to develop such
450
technology capable of installing large MAC/HACs is imperative.
451
In our view, considering these factors and the relative difficulties of MMCT and other non-yeast
452
base MAC/HAC installation technologies, it seems highly likely that YACs delivered to
453
mammalian cells using spheroplast fusion could be developed to produce stable HACs. Yeast
454
chromatin can be successfully expressed, replicated, and segregated in mammalian cells.
455
Cloning alphoid DNA in a yeast vector while including binding sites for histone
456
acetyltransferases and CENP-A or HJURP will likely form stable HACs once delivered into
457
mammalian cells. Using yeast to manipulate HACs would greatly extend the applicability of
458
HACs and may enable HACs capable of being easily constructed, modified, and delivered using
459
yeast vectors.
460
A driver for all of the anticipated MAC/HAC advances will be the potential of this technology for
461
gene therapies, construction of higher eukaryotic cells containing artificial chromosomes
462
encoding pharmaceuticals, the production of new transgenic animals, and likely, although not
463
discussed in this review, transgenic plants containing plant artificial chromosomes. We envision
464
the artificial chromosome technology presented here will evolve to become a major force in the
465
growing bioeconomy.
466
467
Acknowledgements: This work was supported by the Defense Advanced Research Projects
468
Agency [W911NF-11-2-0056]; and the J. Craig Venter Institute. We thank Lauren Perillo-Brown
469
for her diligent proofreading of this article.
470 471
Figure 1. Schematic representation of TAR cloning. In this example, two fragments (red and
472
blue) are PCR amplified with tails containing sequence overlaps to both a vector containing a
473
YAC (black) and to a gene in genomic DNA (green). Yeast can then recombine the overlaps to
474
assemble a desired molecule.
475 476
Figure 2. Schematic representation of yeast fusion to deliver synthetic DNA construct.
477
First, yeast is produced containing the synthetic DNA vector cloned as a YAC or YCp. Next the
478
yeast cell wall is removed by enzymatic digestion to create spheroplasts. PEG is used to fuse
479
the membrane bilayers of the two cell types. The YAC DNA then migrates across the cytoplasm
480
and is taken up by the mammalian nucleus and expressed. Endogenous yeast chromosomes
481
are lost because they have no machinery to drive replication or segregation.
482 483
Figure 3. Diagram of the technologies used to build and transfer MAC/HACs
484
The original MAC/HAC technology, a top-down strategy based on installation of new HAC
485
genetic payloads into existing small chromosomes, and MMCT for MAC/HAC installation is
486
shown on the left.
487
methods made possible by the emergence of synthetic biology and new tools in yeast and
488
bacterial genetics.
489
transferred to DT40 cells. The HAC is then truncated and the insertion of a LoxP site allows for
On the right, newer bottom-up MAC/HAC construction and installation
To construct a top-down HAC, first the full human chromosome is
490
modification and minimization of the HAC and the chromosome can then be transferred to other
491
cell types via MMCT.
492
assembling oligos, ordering genetic cassettes, or amplifying synthetic alphoid DNA via rolling
493
circle amplification. Modifications and assemblies can be made in vitro by homology-based
494
assembly, or by modifying and building HACs in yeast by TAR cloning. Next the bottom-up
495
HAC can be transferred to an animal cell line by transfection or yeast fusion for larger
496
constructs.
To construct a bottom-up HAC, first synthetic DNA is produced by
497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527
1. 2. 3. 4.
5.
6.
7.
8. 9. 10.
Kouprina N, Larionov V. 2016. Transformation-associated recombination (TAR) cloning for genomics studies and synthetic biology. Chromosoma 125:621-32. Khalil AS, Collins JJ. 2010. Synthetic biology: applications come of age. Nat Rev Genet 11:367-79. Lienert F, Lohmueller JJ, Garg A, Silver PA. 2014. Synthetic biology in mammalian cells: next generation research tools and therapeutics. Nat Rev Mol Cell Biol 15:95-107. Gibson DG, Benders GA, Andrews-Pfannkoch C, Denisova EA, Baden-Tillson H, Zaveri J, Stockwell TB, Brownley A, Thomas DW, Algire MA, Merryman C, Young L, Noskov VN, Glass JI, Venter JC, Hutchison CA, 3rd, Smith HO. 2008. Complete chemical synthesis, assembly, and cloning of a Mycoplasma genitalium genome. Science 319:1215-20. Gibson DG, Glass JI, Lartigue C, Noskov VN, Chuang RY, Algire MA, Benders GA, Montague MG, Ma L, Moodie MM, Merryman C, Vashee S, Krishnakumar R, Assad-Garcia N, AndrewsPfannkoch C, Denisova EA, Young L, Qi ZQ, Segall-Shapiro TH, Calvey CH, Parmar PP, Hutchison CA, 3rd, Smith HO, Venter JC. 2010. Creation of a bacterial cell controlled by a chemically synthesized genome. Science 329:52-6. Dormitzer PR, Suphaphiphat P, Gibson DG, Wentworth DE, Stockwell TB, Algire MA, Alperovich N, Barro M, Brown DM, Craig S, Dattilo BM, Denisova EA, De Souza I, Eickmann M, Dugan VG, Ferrari A, Gomila RC, Han L, Judge C, Mane S, Matrosovich M, Merryman C, Palladino G, Palmer GA, Spencer T, Strecker T, Trusheim H, Uhlendorff J, Wen Y, Yee AC, Zaveri J, Zhou B, Becker S, Donabedian A, Mason PW, Glass JI, Rappuoli R, Venter JC. 2013. Synthetic generation of influenza vaccine viruses for rapid response to pandemics. Sci Transl Med 5:185ra68. Gibson DG, Benders GA, Axelrod KC, Zaveri J, Algire MA, Moodie M, Montague MG, Venter JC, Smith HO, Hutchison CA, 3rd. 2008. One-step assembly in yeast of 25 overlapping DNA fragments to form a complete synthetic Mycoplasma genitalium genome. Proc Natl Acad Sci U S A 105:20404-9. Chao R, Yuan Y, Zhao H. 2015. Recent advances in DNA assembly technologies. FEMS Yeast Res 15:1-9. Horton RM, Hunt HD, Ho SN, Pullen JK, Pease LR. 1989. Engineering hybrid genes without the use of restriction enzymes: gene splicing by overlap extension. Gene 77:61-8. Quan J, Tian J. 2009. Circular polymerase extension cloning of complex gene libraries and pathways. PLoS One 4:e6441.
528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575
11. 12. 13.
14. 15.
16. 17. 18. 19.
20.
21. 22.
23.
24. 25.
26.
27.
Li MZ, Elledge SJ. 2007. Harnessing homologous recombination in vitro to generate recombinant DNA via SLIC. Nat Methods 4:251-6. Zhang Y, Werling U, Edelmann W. 2012. SLiCE: a novel bacterial cell extract-based DNA cloning method. Nucleic Acids Res 40:e55. Colloms SD, Merrick CA, Olorunniji FJ, Stark WM, Smith MC, Osbourn A, Keasling JD, Rosser SJ. 2014. Rapid metabolic pathway assembly and modification using serine integrase site-specific recombination. Nucleic Acids Res 42:e23. Nour-Eldin HH, Geu-Flores F, Halkier BA. 2010. USER cloning and USER fusion: the ideal cloning techniques for small and big laboratories. Methods Mol Biol 643:185-200. Wang L, Jiang S, Chen C, He W, Wu X, Wang F, Tong T, Zou X, Li Z, Luo J, Deng Z, Chen S. 2018. Synthetic Genomics: From DNA Synthesis to Genome Design. Angew Chem Int Ed Engl 57:17481756. Smolke CD. 2009. Building outside of the box: iGEM and the BioBricks Foundation. Nat Biotechnol 27:1099-102. Anderson JC, Dueber JE, Leguia M, Wu GC, Goler JA, Arkin AP, Keasling JD. 2010. BglBricks: A flexible standard for biological part assembly. J Biol Eng 4:1. Engler C, Kandzia R, Marillonnet S. 2008. A one pot, one step, precision cloning method with high throughput capability. PLoS One 3:e3647. de Kok S, Stanton LH, Slaby T, Durot M, Holmes VF, Patel KG, Platt D, Shapland EB, Serber Z, Dean J, Newman JD, Chandran SS. 2014. Rapid and reliable DNA assembly via ligase cycling reaction. ACS Synth Biol 3:97-106. Noskov VN, Karas BJ, Young L, Chuang RY, Gibson DG, Lin YC, Stam J, Yonemoto IT, Suzuki Y, Andrews-Pfannkoch C, Glass JI, Smith HO, Hutchison CA, 3rd, Venter JC, Weyman PD. 2012. Assembly of large, high G+C bacterial DNA fragments in yeast. ACS Synth Biol 1:267-73. Gibson DG, Smith HO, Hutchison CA, 3rd, Venter JC, Merryman C. 2010. Chemical synthesis of the mouse mitochondrial genome. Nat Methods 7:901-3. Benders GA, Noskov VN, Denisova EA, Lartigue C, Gibson DG, Assad-Garcia N, Chuang RY, Carrera W, Moodie M, Algire MA, Phan Q, Alperovich N, Vashee S, Merryman C, Venter JC, Smith HO, Glass JI, Hutchison CA, 3rd. 2010. Cloning whole bacterial genomes in yeast. Nucleic Acids Res 38:2558-69. Hutchison CA, 3rd, Chuang RY, Noskov VN, Assad-Garcia N, Deerinck TJ, Ellisman MH, Gill J, Kannan K, Karas BJ, Ma L, Pelletier JF, Qi ZQ, Richter RA, Strychalski EA, Sun L, Suzuki Y, Tsvetanova B, Wise KS, Smith HO, Glass JI, Merryman C, Gibson DG, Venter JC. 2016. Design and synthesis of a minimal bacterial genome. Science 351:aad6253. Richardson SM, Nunley PW, Yarrington RM, Boeke JD, Bader JS. 2010. GeneDesign 3.0 is an updated synthetic biology toolkit. Nucleic Acids Res 38:2603-6. Richardson SM, Mitchell LA, Stracquadanio G, Yang K, Dymond JS, DiCarlo JE, Lee D, Huang CL, Chandrasegaran S, Cai Y, Boeke JD, Bader JS. 2017. Design of a synthetic yeast genome. Science 355:1040-1044. Shen Y, Wang Y, Chen T, Gao F, Gong J, Abramczyk D, Walker R, Zhao H, Chen S, Liu W, Luo Y, Muller CA, Paul-Dubois-Taine A, Alver B, Stracquadanio G, Mitchell LA, Luo Z, Fan Y, Zhou B, Wen B, Tan F, Wang Y, Zi J, Xie Z, Li B, Yang K, Richardson SM, Jiang H, French CE, Nieduszynski CA, Koszul R, Marston AL, Yuan Y, Wang J, Bader JS, Dai J, Boeke JD, Xu X, Cai Y, Yang H. 2017. Deep functional analysis of synII, a 770-kilobase synthetic yeast chromosome. Science 355. Mitchell LA, Wang A, Stracquadanio G, Kuang Z, Wang X, Yang K, Richardson S, Martin JA, Zhao Y, Walker R, Luo Y, Dai H, Dong K, Tang Z, Yang Y, Cai Y, Heguy A, Ueberheide B, Fenyo D, Dai J, Bader JS, Boeke JD. 2017. Synthesis, debugging, and effects of synthetic chromosome consolidation: synVI and beyond. Science 355.
576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621
28.
29. 30. 31.
32. 33. 34.
35.
36.
37. 38.
39.
40.
41.
42.
43.
Zhao Y, Boeke JD. 2018. Construction of Designer Selectable Marker Deletions with a CRISPRCas9 Toolbox in Schizosaccharomyces pombe and New Design of Common Entry Vectors. G3 (Bethesda) 8:789-796. Pretorius IS, Boeke JD. 2018. Yeast 2.0-connecting the dots in the construction of the world's first functional synthetic eukaryotic genome. FEMS Yeast Res 18. Luo J, Sun X, Cormack BP, Boeke JD. 2018. Karyotype engineering by chromosome fusion leads to reproductive isolation in yeast. Nature 560:392-396. Wu Y, Zhu RY, Mitchell LA, Ma L, Liu R, Zhao M, Jia B, Xu H, Li YX, Yang ZM, Ma Y, Li X, Liu H, Liu D, Xiao WH, Zhou X, Li BZ, Yuan YJ, Boeke JD. 2018. In vitro DNA SCRaMbLE. Nat Commun 9:1935. Luo Z, Wang L, Wang Y, Zhang W, Guo Y, Shen Y, Jiang L, Wu Q, Zhang C, Cai Y, Dai J. 2018. Identifying and characterizing SCRaMbLEd synthetic yeast using ReSCuES. Nat Commun 9:1930. Szymanski E, Calvert J. 2018. Designing with living systems in the synthetic yeast project. Nat Commun 9:2950. Noskov VN, Segall-Shapiro TH, Chuang RY. 2010. Tandem repeat coupled with endonuclease cleavage (TREC): a seamless modification tool for genome engineering in yeast. Nucleic Acids Res 38:2570-6. Chandran S, Noskov VN, Segall-Shapiro TH, Ma L, Whiteis C, Lartigue C, Jores J, Vashee S, Chuang RY. 2014. TREC-IN: gene knock-in genetic tool for genomes cloned in yeast. BMC Genomics 15:1180. Oldfield LM, Grzesik P, Voorhies AA, Alperovich N, MacMath D, Najera CD, Chandra DS, Prasad S, Noskov VN, Montague MG, Friedman RM, Desai PJ, Vashee S. 2017. Genome-wide engineering of an infectious clone of herpes simplex virus type 1 using synthetic genomics assembly methods. Proc Natl Acad Sci U S A 114:E8885-E8894. Gibson DG. 2009. Synthesis of DNA fragments in yeast by one-step assembly of overlapping oligonucleotides. Nucleic Acids Res 37:6984-90. Kouprina N, Ebersole T, Koriabine M, Pak E, Rogozin IB, Katoh M, Oshimura M, Ogi K, Peredelchuk M, Solomon G, Brown W, Barrett JC, Larionov V. 2003. Cloning of human centromeres by transformation-associated recombination in yeast and generation of functional human artificial chromosomes. Nucleic Acids Res 31:922-34. Ebersole T, Okamoto Y, Noskov VN, Kouprina N, Kim JH, Leem SH, Barrett JC, Masumoto H, Larionov V. 2005. Rapid generation of long synthetic tandem repeats and its application for analysis in human artificial chromosome formation. Nucleic Acids Res 33:e130. Kim JH, Kononenko A, Erliandri I, Kim TA, Nakano M, Iida Y, Barrett JC, Oshimura M, Masumoto H, Earnshaw WC, Larionov V, Kouprina N. 2011. Human artificial chromosome (HAC) vector with a conditional centromere for correction of genetic deficiencies in human cells. Proc Natl Acad Sci U S A 108:20048-53. Iida Y, Kim JH, Kazuki Y, Hoshiya H, Takiguchi M, Hayashi M, Erliandri I, Lee HS, Samoshkin A, Masumoto H, Earnshaw WC, Kouprina N, Larionov V, Oshimura M. 2010. Human artificial chromosome with a conditional centromere for gene delivery and gene expression. DNA Res 17:293-301. Kononenko AV, Bansal R, Lee NC, Grimes BR, Masumoto H, Earnshaw WC, Larionov V, Kouprina N. 2014. A portable BRCA1-HAC (human artificial chromosome) module for analysis of BRCA1 tumor suppressor function. Nucleic Acids Res 42:e164-e164. Ohzeki J, Larionov V, Earnshaw WC, Masumoto H. 2019. De novo formation and epigenetic maintenance of centromere chromatin. Curr Opin Cell Biol 58:15-25.
622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668
44.
45. 46. 47. 48. 49.
50.
51. 52.
53.
54.
55. 56.
57.
58. 59.
60. 61. 62.
Bergmann JH, Martins NM, Larionov V, Masumoto H, Earnshaw WC. 2012. HACking the centromere chromatin code: insights from human artificial chromosomes. Chromosome Res 20:505-19. Stellfox ME, Bailey AO, Foltz DR. 2013. Putting CENP-A in its place. Cell Mol Life Sci 70:387-406. Ohzeki J, Larionov V, Earnshaw WC, Masumoto H. 2015. Genetic and epigenetic regulation of centromeres: a look at HAC formation. Chromosome Res 23:87-103. Pesenti ME, Weir JR, Musacchio A. 2016. Progress in the structural and functional characterization of kinetochores. Curr Opin Struct Biol 37:152-63. Schalch T, Steiner FA. 2017. Structure of centromere chromatin: from nucleosome to chromosomal architecture. Chromosoma 126:443-455. Waye JS, Durfy SJ, Pinkel D, Kenwrick S, Patterson M, Davies KE, Willard HF. 1987. Chromosomespecific alpha satellite DNA from human chromosome 1: hierarchical structure and genomic organization of a polymorphic domain spanning several hundred kilobase pairs of centromeric DNA. Genomics 1:43-51. Swartz SZ, McKay LS, Su KC, Bury L, Padeganeh A, Maddox PS, Knouse KA, Cheeseman IM. 2019. Quiescent Cells Actively Replenish CENP-A Nucleosomes to Maintain Centromere Identity and Proliferative Potential. Dev Cell 51:35-48 e7. Grimes BR, Schindelhauer D, McGill NI, Ross A, Ebersole TA, Cooke HJ. 2001. Stable gene expression from a mammalian artificial chromosome. EMBO Rep 2:910-4. Ohzeki J, Bergmann JH, Kouprina N, Noskov VN, Nakano M, Kimura H, Earnshaw WC, Larionov V, Masumoto H. 2012. Breaking the HAC Barrier: histone H3K9 acetyl/methyl balance regulates CENP-A assembly. EMBO J 31:2391-402. Masumoto H, Masukata H, Muro Y, Nozaki N, Okazaki T. 1989. A human centromere antigen (CENP-B) interacts with a short specific sequence in alphoid DNA, a human centromeric satellite. J Cell Biol 109:1963-73. Dunleavy EM, Roche D, Tagami H, Lacoste N, Ray-Gallet D, Nakamura Y, Daigo Y, Nakatani Y, Almouzni-Pettinotti G. 2009. HJURP is a cell-cycle-dependent maintenance and deposition factor of CENP-A at centromeres. Cell 137:485-97. Foltz DR, Jansen LE, Bailey AO, Yates JR, 3rd, Bassett EA, Wood S, Black BE, Cleveland DW. 2009. Centromere-specific assembly of CENP-a nucleosomes is mediated by HJURP. Cell 137:472-84. Barnhart MC, Kuich PH, Stellfox ME, Ward JA, Bassett EA, Black BE, Foltz DR. 2011. HJURP is a CENP-A chromatin assembly factor sufficient to form a functional de novo kinetochore. J Cell Biol 194:229-43. Logsdon GA, Barrey EJ, Bassett EA, DeNizio JE, Guo LY, Panchenko T, Dawicki-McKenna JM, Heun P, Black BE. 2015. Both tails and the centromere targeting domain of CENP-A are required for centromere establishment. J Cell Biol 208:521-31. Talbert PB, Henikoff S. 2006. Spreading of silent chromatin: inaction at a distance. Nat Rev Genet 7:793-803. Fachinetti D, Folco HD, Nechemia-Arbely Y, Valente LP, Nguyen K, Wong AJ, Zhu Q, Holland AJ, Desai A, Jansen LE, Cleveland DW. 2013. A two-step mechanism for epigenetic specification of centromere identity and function. Nat Cell Biol 15:1056-66. Martella A, Pollard SM, Dai J, Cai Y. 2016. Mammalian Synthetic Biology: Time for Big MACs. ACS Synth Biol 5:1040-1049. Kazuki Y, Oshimura M. 2011. Human artificial chromosomes for gene delivery and the development of animal models. Mol Ther 19:1591-601. Kouprina N, Earnshaw WC, Masumoto H, Larionov V. 2013. A new generation of human artificial chromosomes for functional genomics and gene therapy. Cell Mol Life Sci 70:1135-48.
669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715
63.
64.
65.
66. 67.
68.
69.
70. 71. 72.
73.
74.
75.
76. 77. 78.
79.
Oshimura M, Uno N, Kazuki Y, Katoh M, Inoue T. 2015. A pathway from chromosome transfer to engineering resulting in human and mouse artificial chromosomes for a variety of applications to bio-medical challenges. Chromosome Res 23:111-33. Katoh M, Ayabe F, Norikane S, Okada T, Masumoto H, Horike S, Shirayoshi Y, Oshimura M. 2004. Construction of a novel human artificial chromosome vector for gene delivery. Biochem Biophys Res Commun 321:280-90. Kazuki Y, Hoshiya H, Takiguchi M, Abe S, Iida Y, Osaki M, Katoh M, Hiratsuka M, Shirayoshi Y, Hiramatsu K, Ueno E, Kajitani N, Yoshino T, Kazuki K, Ishihara C, Takehara S, Tsuji S, Ejima F, Toyoda A, Sakaki Y, Larionov V, Kouprina N, Oshimura M. 2011. Refined human artificial chromosome vectors for gene therapy and animal transgenesis. Gene Ther 18:384-93. Heller R, Brown KE, Burgtorf C, Brown WR. 1996. Mini-chromosomes derived from the human Y chromosome by telomere directed chromosome breakage. Proc Natl Acad Sci U S A 93:7125-30. Kakeda M, Nagata K, Osawa K, Matsuno H, Hiratsuka M, Sano A, Okazaki A, Shitara S, Nishikawa S, Masuya A, Hata T, Wako S, Osaki M, Kazuki Y, Oshimura M, Tomizuka K. 2011. A new chromosome 14-based human artificial chromosome (HAC) vector system for efficient transgene expression in human primary cells. Biochem Biophys Res Commun 415:439-44. Mills W, Critcher R, Lee C, Farr CJ. 1999. Generation of an approximately 2.4 Mb human X centromere-based minichromosome by targeted telomere-associated chromosome fragmentation in DT40. Hum Mol Genet 8:751-61. Harrington JJ, Van Bokkelen G, Mays RW, Gustashaw K, Willard HF. 1997. Formation of de novo centromeres and construction of first-generation human artificial microchromosomes. Nat Genet 15:345-55. Ikeno M, Grimes B, Okazaki T, Nakano M, Saitoh K, Hoshino H, McGill NI, Cooke H, Masumoto H. 1998. Construction of YAC-based mammalian artificial chromosomes. Nat Biotechnol 16:431-9. Logsdon GA, Gambogi CW, Liskovykh MA, Barrey EJ, Larionov V, Miga KH, Heun P, Black BE. 2019. Human Artificial Chromosomes that Bypass Centromeric DNA. Cell 178:624-639 e19. Kouprina N, Petrov N, Molina O, Liskovykh M, Pesenti E, Ohzeki JI, Masumoto H, Earnshaw WC, Larionov V. 2018. Human Artificial Chromosome with Regulated Centromere: A Tool for Genome and Cancer Studies. ACS Synth Biol 7:1974-1989. Kouprina N, Samoshkin A, Erliandri I, Nakano M, Lee HS, Fu H, Iida Y, Aladjem M, Oshimura M, Masumoto H, Earnshaw WC, Larionov V. 2012. Organization of synthetic alphoid DNA array in human artificial chromosome (HAC) with a conditional centromere. ACS Synth Biol 1:590-601. Zhu Z, Ma B, Homer RJ, Zheng T, Elias JA. 2001. Use of the tetracycline-controlled transcriptional silencer (tTS) to eliminate transgene leak in inducible overexpression transgenic mice. J Biol Chem 276:25222-9. Shono N, Ohzeki J, Otake K, Martins NM, Nagase T, Kimura H, Larionov V, Earnshaw WC, Masumoto H. 2015. CENP-C and CENP-I are key connecting factors for kinetochore and CENP-A assembly. J Cell Sci 128:4572-87. Ohzeki Ji. 2002. CENP-B box is required for de novo centromere chromatin assembly on human alphoid DNA. The Journal of Cell Biology 159:765-775. Nagpal H, Fukagawa T. 2016. Kinetochore assembly and function through the cell cycle. Chromosoma 125:645-59. Brown DM, Chan YA, Desai PJ, Grzesik P, Oldfield LM, Vashee S, Way JC, Silver PA, Glass JI. 2017. Efficient size-independent chromosome delivery from yeast to cultured cell lines. Nucleic Acids Res 45:e50. Lartigue C, Vashee S, Algire MA, Chuang RY, Benders GA, Ma L, Noskov VN, Denisova EA, Gibson DG, Assad-Garcia N, Alperovich N, Thomas DW, Merryman C, Hutchison CA, 3rd, Smith HO,
716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762
80.
81.
82. 83.
84.
85.
86. 87. 88. 89. 90. 91.
92. 93.
94.
95. 96. 97. 98.
Venter JC, Glass JI. 2009. Creating bacterial strains from genomes that have been cloned and engineered in yeast. Science 325:1693-6. Karas BJ, Molparia B, Jablanovic J, Hermann WJ, Lin YC, Dupont CL, Tagwerker C, Yonemoto IT, Noskov VN, Chuang RY, Allen AE, Glass JI, Hutchison CA, 3rd, Smith HO, Venter JC, Weyman PD. 2013. Assembly of eukaryotic algal chromosomes in yeast. J Biol Eng 7:30. Zhang XF, Wu GX, Chen JQ, Zhang AM, Liu SG, Jiao BH, Cheng GX. 2005. Transfer of an expression YAC into goat fetal fibroblasts by cell fusion for mammary gland bioreactor. Biochem Biophys Res Commun 333:58-63. Li L, Blankenstein T. 2013. Generation of transgenic mice with megabase-sized human yeast artificial chromosomes by yeast spheroplast-embryonic stem cell fusion. Nat Protoc 8:1567-82. Huxley C, Hagino Y, Schlessinger D, Olson MV. 1991. The Human HPRT Gene on a Yeast Artificial Chromosome Is Functional When Transferred to Mouse Cells by Cell Fusion. Genomics 9:742750. Pachnis V, Pevny L, Rothstein R, Costantini F. 1990. Transfer of a yeast artificial chromosome carrying human DNA from Saccharomyces cerevisiae into mammalian cells. Proc Natl Acad Sci U S A 87:5109-13. Julicher K, Vieten L, Brocker F, Bardenheuer W, Schutte J, Opalka B. 1997. Yeast artificial chromosome transfer into human renal carcinoma cells by spheroplast fusion. Genomics 43:958. Simpson K, McGuigan A, Huxley C. 1996. Stable episomal maintenance of yeast artificial chromosomes in human cells. Mol Cell Biol 16:5117-26. Simpson K, Huxley C. 1996. A shuttle system for transfer of YACs between yeast and mammalian cells. Nucleic Acids Res 24:4693-9. Auriche C, Carpani D, Conese M, Caci E, Zegarra-Moran O, Donini P, Ascenzioni F. 2002. Functional human CFTR produced by a stable minichromosome. EMBO Rep 3:862-8. Marschall P, Malik N, Larin Z. 1999. Transfer of YACs up to 2.3 Mb intact into human cells with polyethylenimine. Gene Ther 6:1634-7. Davies NP, Rosewell IR, Bruggemann M. 1992. Targeted alterations in yeast artificial chromosomes for inter-species gene transfer. Nucleic Acids Res 20:2693-8. Rocchi L, Braz C, Cattani S, Ramalho A, Christan S, Edlinger M, Ascenzioni F, Laner A, Kraner S, Amaral M, Schindelhauer D. 2010. Escherichia coli-cloned CFTR loci relevant for human artificial chromosome therapy. Hum Gene Ther 21:1077-92. Bauchwitz R, Costantini F. 1998. YAC transgenesis: a study of conditions to protect YAC DNA from breakage and a protocol for transfection. Biochim Biophys Acta 1401:21-37. Karas BJ, Jablanovic J, Irvine E, Sun L, Ma L, Weyman PD, Gibson DG, Glass JI, Venter JC, Hutchison CA, 3rd, Smith HO, Suzuki Y. 2014. Transferring whole genomes from bacteria to yeast spheroplasts using entire bacterial cells to reduce DNA shearing. Nat Protoc 9:743-50. Shao Y, Lu N, Wu Z, Cai C, Wang S, Zhang LL, Zhou F, Xiao S, Liu L, Zeng X, Zheng H, Yang C, Zhao Z, Zhao G, Zhou JQ, Xue X, Qin Z. 2018. Creating a functional single-chromosome yeast. Nature 560:331-335. Cevc G, Richardsen H. 1999. Lipid vesicles and membrane fusion. Adv Drug Deliv Rev 38:207232. Tamm LK, Crane J, Kiessling V. 2003. Membrane fusion: a structural perspective on the interplay of lipids and proteins. Curr Opin Struct Biol 13:453-66. Lentz BR. 2007. PEG as a tool to gain insight into membrane fusion. Eur Biophys J 36:315-26. Saez R, Alonso A, Villena A, Goni FM. 1982. Detergent-like properties of polyethyleneglycols in relation to model membranes. FEBS Lett 137:323-6.
763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808
99. 100.
101. 102.
103. 104. 105. 106.
107. 108.
109. 110. 111. 112. 113. 114. 115. 116.
117.
118.
Boni LT, Stewart TP, Hui SW. 1984. Alterations in phospholipid polymorphism by polyethylene glycol. J Membr Biol 80:91-104. Maehara T, Itaya M, Ogura M, Tanaka T. 2011. Effect of Bacillus subtilis BsuM restrictionmodification on plasmid transfer by polyethylene glycol-induced protoplast fusion. FEMS Microbiol Lett 325:49-55. Baigori M, Sesma F, Holgado APD, Demendoza D. 1988. Transfer of plasmids between bacillussubtilis and streptococcus-lactis. Applied and Environmental Microbiology 54:1309-1311. Dai M, Ziesman S, Ratcliffe T, Gill RT, Copley SD. 2005. Visualization of protoplast fusion and quantitation of recombination in fused protoplasts of auxotrophic strains of Escherichia coli. Metab Eng 7:45-52. Tarshis M, Salman M, Rottem S. 1991. Fusion of mycoplasmas: the formation of cell hybrids. FEMS Microbiol Lett 66:67-71. Lartigue C, Glass JI, Alperovich N, Pieper R, Parmar PP, Hutchison CA, 3rd, Smith HO, Venter JC. 2007. Genome transplantation in bacteria: changing one species to another. Science 317:632-8. Gyuris J, Duda EG. 1986. High-efficiency transformation of Saccharomyces cerevisiae cells by bacterial minicell protoplast fusion. Mol Cell Biol 6:3295-7. Guerratschuschke I, Martin I, Gonzalez MT. 1991. Polyethylene glycol-induced internalization of bacteria into fungal protoplasts: electron microscopic study and optimization of experimental conditions. Applied and Environmental Microbiology 57:1516-1522. Schaffner W. 1980. Direct transfer of cloned genes from bacteria to mammalian cells. Proc Natl Acad Sci U S A 77:2163-7. Karas BJ, Jablanovic J, Sun L, Ma L, Goldgof GM, Stam J, Ramon A, Manary MJ, Winzeler EA, Venter JC, Weyman PD, Gibson DG, Glass JI, Hutchison CA, 3rd, Smith HO, Suzuki Y. 2013. Direct transfer of whole genomes from bacteria to yeast. Nat Methods 10:410-2. Vansolingen P, Vanderplaat JB. 1977. Fusion of yeast spheroplasts. Journal of Bacteriology 130:946-947. Markie D. 1996. A simple assay for optimizing yeast-mammalian cell fusion conditions. Mol Biotechnol 6:99-104. Fournier RE, Ruddle FH. 1977. Microcell-mediated transfer of murine chromosomes into mouse, Chinese hamster, and human somatic cells. Proc Natl Acad Sci U S A 74:319-23. Ege T, Ringertz NR. 1974. Preparation of microcells by enucleation of micronucleate cells. Exp Cell Res 87:378-82. Meaburn KJ, Parris CN, Bridger JM. 2005. The manipulation of chromosomes by mankind: the uses of microcell-mediated chromosome transfer. Chromosoma 114:263-74. Schor SL, Johnson RT, Mullinger AM. 1975. Perturbation of mammalian cell division. II. Studies on the isolation and characterization of human mini segregant cells. J Cell Sci 19:281-303. Liskovykh M, Lee NC, Larionov V, Kouprina N. 2016. Moving toward a higher efficiency of microcell-mediated chromosome transfer. Mol Ther Methods Clin Dev 3:16043. Suzuki T, Kazuki Y, Oshimura M, Hara T. 2016. Highly Efficient Transfer of Chromosomes to a Broad Range of Target Cells Using Chinese Hamster Ovary Cells Expressing Murine Leukemia Virus-Derived Envelope Proteins. PLoS One 11:e0157187. Hiratsuka M, Ueda K, Uno N, Uno K, Fukuhara S, Kurosaki H, Takehara S, Osaki M, Kazuki Y, Kurosawa Y, Nakamura T, Katoh M, Oshimura M. 2015. Retargeting of microcell fusion towards recipient cell-oriented transfer of human artificial chromosome. BMC Biotechnol 15:58. Doherty AM, Fisher EM. 2003. Microcell-mediated chromosome transfer (MMCT): small cells with huge potential. Mamm Genome 14:583-92.
809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854
119.
120.
121.
122. 123. 124.
125. 126. 127. 128. 129. 130.
131. 132. 133.
134.
135.
136.
Tomizuka K, Yoshida H, Uejima H, Kugoh H, Sato K, Ohguma A, Hayasaka M, Hanaoka K, Oshimura M, Ishida I. 1997. Functional expression and germline transmission of a human chromosome fragment in chimaeric mice. Nat Genet 16:133-43. Yoshimura Y, Nakamura K, Endo T, Kajitani N, Kazuki K, Kazuki Y, Kugoh H, Oshimura M, Ohbayashi T. 2015. Mouse embryonic stem cells with a multi-integrase mouse artificial chromosome for transchromosomic mouse generation. Transgenic Res 24:717-27. Uno N, Abe S, Oshimura M, Kazuki Y. 2018. Combinations of chromosome transfer and genome editing for the development of cell/animal models of human disease and humanized animal models. J Hum Genet 63:145-156. Capecchi MR. 1980. High efficiency transformation by direct microinjection of DNA into cultured mammalian cells. Cell 22:479-488. Gnirke A, Huxley C, Peterson K, Olson MV. 1993. Microinjection of intact 200- to 500-kb fragments of YAC DNA into mammalian cells. Genomics 15:659-67. Co DO, Borowski AH, Leung JD, van der Kaa J, Hengst S, Platenburg GJ, Pieper FR, Perez CF, Jirik FR, Drayer JI. 2000. Generation of transgenic mice and germline transmission of a mammalian artificial chromosome introduced into embryos by pronuclear microinjection. Chromosome Res 8:183-91. Kagawa Y, Inoki Y, Endo H. 2001. Gene therapy by mitochondrial transfer. Adv Drug Deliv Rev 49:107-19. Hoess RH, Abremski K. 1984. Interaction of the bacteriophage P1 recombinase Cre with the recombining site loxP. Proc Natl Acad Sci U S A 81:1026-9. Sauer B, Henderson N. 1988. Site-specific DNA recombination in mammalian cells by the Cre recombinase of bacteriophage P1. Proc Natl Acad Sci U S A 85:5166-70. Guo F, Gopaul DN, van Duyne GD. 1997. Structure of Cre recombinase complexed with DNA in a site-specific recombination synapse. Nature 389:40-6. Sauer B. 1987. Functional expression of the cre-lox site-specific recombination system in the yeast Saccharomyces cerevisiae. Mol Cell Biol 7:2087-96. Turan S, Galla M, Ernst E, Qiao J, Voelkel C, Schiedlmeier B, Zehe C, Bode J. 2011. Recombinasemediated cassette exchange (RMCE): traditional concepts and current challenges. J Mol Biol 407:193-221. Turan S, Qiao J, Madden S, Benham C, Kotz M, Schambach A, Bode J. 2014. Expanding Flp-RMCE options: the potential of Recombinase Mediated Twin-Site Targeting (RMTT). Gene 546:135-44. Osterwalder M, Galli A, Rosen B, Skarnes WC, Zeller R, Lopez-Rios J. 2010. Dual RMCE for efficient re-engineering of mouse mutant alleles. Nat Methods 7:893-5. Callesen MM, Berthelsen MF, Lund S, Fuchtbauer AC, Fuchtbauer EM, Jakobsen JE. 2016. Recombinase-Mediated Cassette Exchange (RMCE)-in Reporter Cell Lines as an Alternative to the Flp-in System. PLoS One 11:e0161471. Kannan K, Tsvetanova B, Chuang RY, Noskov VN, Assad-Garcia N, Ma L, Hutchison Iii CA, Smith HO, Glass JI, Merryman C, Venter JC, Gibson DG. 2016. One step engineering of the smallsubunit ribosomal RNA using CRISPR/Cas9. Sci Rep 6:30714. Ruiz E, Talenton V, Dubrana MP, Guesdon G, Lluch-Senar M, Salin F, Sirand-Pugnet P, Arfi Y, Lartigue C. 2019. CReasPy-Cloning: A Method for Simultaneous Cloning and Engineering of Megabase-Sized Genomes in Yeast Using the CRISPR-Cas9 System. ACS Synth Biol doi:10.1021/acssynbio.9b00224. DiCarlo JE, Norville JE, Mali P, Rios X, Aach J, Church GM. 2013. Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res 41:4336-43.
855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900
137.
138. 139.
140. 141.
142. 143. 144.
145.
146. 147.
148.
149. 150. 151. 152. 153. 154.
155.
Xie ZX, Mitchell LA, Liu HM, Li BZ, Liu D, Agmon N, Wu Y, Li X, Zhou X, Li B, Xiao WH, Ding MZ, Wang Y, Yuan YJ, Boeke JD. 2018. Rapid and Efficient CRISPR/Cas9-Based Mating-Type Switching of Saccharomyces cerevisiae. G3 (Bethesda) 8:173-183. Generoso WC, Gottardi M, Oreb M, Boles E. 2016. Simplified CRISPR-Cas genome editing for Saccharomyces cerevisiae. J Microbiol Methods 127:203-205. Jakociunas T, Bonde I, Herrgard M, Harrison SJ, Kristensen M, Pedersen LE, Jensen MK, Keasling JD. 2015. Multiplex metabolic pathway engineering using CRISPR/Cas9 in Saccharomyces cerevisiae. Metab Eng 28:213-222. Tsarmpopoulos I, Gourgues G, Blanchard A, Vashee S, Jores J, Lartigue C, Sirand-Pugnet P. 2016. In-Yeast Engineering of a Bacterial Genome Using CRISPR/Cas9. ACS Synth Biol 5:104-9. Sonoda E, Hochegger H, Saberi A, Taniguchi Y, Takeda S. 2006. Differential usage of nonhomologous end-joining and homologous recombination in double strand break repair. DNA Repair (Amst) 5:1021-9. Lorenz MC, Muir RS, Lim E, McElver J, Weber SC, Heitman J. 1995. Gene disruption with PCR products in Saccharomyces cerevisiae. Gene 158:113-7. Baudin A, Ozier-Kalogeropoulos O, Denouel A, Lacroute F, Cullin C. 1993. A simple and efficient method for direct gene deletion in Saccharomyces cerevisiae. Nucleic Acids Res 21:3329-30. Clikeman JA, Khalsa GJ, Barton SL, Nickoloff JA. 2001. Homologous recombinational repair of double-strand breaks in yeast is enhanced by MAT heterozygosity through yKU-dependent and independent mechanisms. Genetics 157:579-89. Kuroiwa Y, Tomizuka K, Shinohara T, Kazuki Y, Yoshida H, Ohguma A, Yamamoto T, Tanaka S, Oshimura M, Ishida I. 2000. Manipulation of human minichromosomes to carry greater than megabase-sized chromosome inserts. Nat Biotechnol 18:1086-90. Spence JM, Mills W, Mann K, Huxley C, Farr CJ. 2006. Increased missegregation and chromosome loss with decreasing chromosome size in vertebrate cells. Chromosoma 115:60-74. Uno N, Hiramatsu K, Uno K, Komoto S, Kazuki Y, Oshimura M. 2017. CRISPR/Cas9-induced transgene insertion and telomere-associated truncation of a single human chromosome for chromosome engineering in CHO and A9 cells. Sci Rep 7:12739. Hoshiya H, Kazuki Y, Abe S, Takiguchi M, Kajitani N, Watanabe Y, Yoshino T, Shirayoshi Y, Higaki K, Messina G, Cossu G, Oshimura M. 2009. A highly stable and nonintegrated human artificial chromosome (HAC) containing the 2.4 Mb entire human dystrophin gene. Mol Ther 17:309-17. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339:819-23. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM. 2013. RNA-guided human genome engineering via Cas9. Science 339:823-6. Cho SW, Kim S, Kim JM, Kim JS. 2013. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol 31:230-2. Hsu PD, Lander ES, Zhang F. 2014. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157:1262-78. Kang Y, Chu C, Wang F, Niu Y. 2019. CRISPR/Cas9-mediated genome editing in nonhuman primates. Dis Model Mech 12. Honma K, Abe S, Endo T, Uno N, Oshimura M, Ohbayashi T, Kazuki Y. 2018. Development of a multiple-gene-loading method by combining multi-integration system-equipped mouse artificial chromosome vector and CRISPR-Cas9. PLoS One 13:e0193642. Gaj T, Gersbach CA, Barbas CF, 3rd. 2013. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 31:397-405.
901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948
156.
157.
158. 159.
160. 161. 162.
163.
164. 165. 166. 167. 168. 169. 170.
171.
172.
173.
174.
Lillico SG, Proudfoot C, Carlson DF, Stverakova D, Neil C, Blain C, King TJ, Ritchie WA, Tan W, Mileham AJ, McLaren DG, Fahrenkrug SC, Whitelaw CB. 2013. Live pigs produced from genome edited zygotes. Sci Rep 3:2847. Carlson DF, Tan W, Lillico SG, Stverakova D, Proudfoot C, Christian M, Voytas DF, Long CR, Whitelaw CB, Fahrenkrug SC. 2012. Efficient TALEN-mediated gene knockout in livestock. Proc Natl Acad Sci U S A 109:17382-7. Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-76. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin, II, Thomson JA. 2007. Induced pluripotent stem cell lines derived from human somatic cells. Science 318:1917-20. Hockemeyer D, Jaenisch R. 2016. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 18:573-86. Aydin B, Mazzoni EO. 2019. Cell Reprogramming: The Many Roads to Success. Annu Rev Cell Dev Biol 35:433-452. Sinenko SA, Skvortsova EV, Liskovykh MA, Ponomartsev SV, Kuzmin AA, Khudiakov AA, Malashicheva AB, Alenina N, Larionov V, Kouprina N, Tomilin AN. 2018. Transfer of Synthetic Human Chromosome into Human Induced Pluripotent Stem Cells for Biomedical Applications. Cells 7. Hiratsuka M, Uno N, Ueda K, Kurosaki H, Imaoka N, Kazuki K, Ueno E, Akakura Y, Katoh M, Osaki M, Kazuki Y, Nakagawa M, Yamanaka S, Oshimura M. 2011. Integration-free iPS cells engineered using human artificial chromosome vectors. PLoS One 6:e25961. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, Wernig M. 2010. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463:1035-41. Li J, Liu X, Wang H, Zhang S, Liu F, Wang X, Wang Y. 2009. Human embryos derived by somatic cell nuclear transfer using an alternative enucleation approach. Cloning Stem Cells 11:39-50. Long CR, Westhusin ME, Golding MC. 2014. Reshaping the transcriptional frontier: epigenetics and somatic cell nuclear transfer. Mol Reprod Dev 81:183-93. Keefer CL. 2015. Artificial cloning of domestic animals. Proc Natl Acad Sci U S A 112:8874-8. Porteus M. 2016. Genome Editing: A New Approach to Human Therapeutics. Annu Rev Pharmacol Toxicol 56:163-90. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. 1997. Viable offspring derived from fetal and adult mammalian cells. Nature 385:810-3. Shinohara T, Kazuki K, Ogonuki N, Morimoto H, Matoba S, Hiramatsu K, Honma K, Suzuki T, Hara T, Ogura A, Oshimura M, Kanatsu-Shinohara M, Kazuki Y. 2017. Transfer of a Mouse Artificial Chromosome into Spermatogonial Stem Cells Generates Transchromosomic Mice. Stem Cell Reports 9:1180-1191. Wu H, Fan Z, Brandsrud M, Meng Q, Bobbitt M, Regouski M, Stott R, Sweat A, Crabtree J, Hogan RJ, Tripp RA, Wang Z, Polejaeva IA, Sullivan EJ. 2019. Generation of H7N9-specific human polyclonal antibodies from a transchromosomic goat (caprine) system. Sci Rep 9:366. Cardenas WB, Loo YM, Gale M, Jr., Hartman AL, Kimberlin CR, Martinez-Sobrido L, Saphire EO, Basler CF. 2006. Ebola virus VP35 protein binds double-stranded RNA and inhibits alpha/beta interferon production induced by RIG-I signaling. J Virol 80:5168-78. Basler CF, Mikulasova A, Martinez-Sobrido L, Paragas J, Muhlberger E, Bray M, Klenk HD, Palese P, Garcia-Sastre A. 2003. The Ebola virus VP35 protein inhibits activation of interferon regulatory factor 3. J Virol 77:7945-56. Boeke JD, Church G, Hessel A, Kelley NJ, Arkin A, Cai Y, Carlson R, Chakravarti A, Cornish VW, Holt L, Isaacs FJ, Kuiken T, Lajoie M, Lessor T, Lunshof J, Maurano MT, Mitchell LA, Rine J, Rosser
949 950 951 952 953
175.
S, Sanjana NE, Silver PA, Valle D, Wang H, Way JC, Yang L. 2016. GENOME ENGINEERING. The Genome Project-Write. Science 353:126-7. Consortium EBR. 2019. A Research Roadmap for the Next-Generation BioEconomy. Consortium EBR, Emeryville, CA.