ArticlePDF Available

Regulated expression of a diphtheria toxin A-chain gene transfected into human cells: Possible strategy for inducing cancer cell suicide

Authors:

Abstract and Figures

As an alternative to directing plant or bacterial toxins to surface receptors, we are investigating the possibility of killing tumor cells by the expression of an exogenously introduced toxin gene (i.e., cell suicide). Tissue-specific gene regulatory elements might thus be exploited to achieve selective killing. To assess the feasibility of such an approach, we have transfected human cells (HeLa, B-lymphoblastoid, and 293 cells) with plasmids containing the diphtheria toxin A-chain (DT-A) coding sequence. The presence of the DT-A sequence lowered the level of transient expression of chloramphenicol acetyltransferase from a cotransfected plasmid, pSV2cat. This expression level in B-cells was further diminished by the inclusion of an immunoglobulin enhancer in the DT-A plasmid. In cotransfection experiments with a DT-A plasmid lacking an enhancer, chloramphenicol acetyltransferase expression was much more strongly inhibited in 293 cells (which express adenovirus E1A and E1B products) than in the other cell types; furthermore, the presence of the DT-A sequence eliminated recovery of G418-resistant 293 cell transformants after transfection with a plasmid containing the neo selectable marker. These results suggest that cell-specific regulatory mechanisms can be exploited to achieve selective cell killing by expression of an introduced toxin gene.
Content may be subject to copyright.
1986;46:4660-4664. Published online September 1, 1986.Cancer Res
Ian H. Maxwell, Françoise Maxwell and L. Michael Glode
Cancer Cell Suicide
Transfected into Human Cells: Possible Strategy for Inducing
Regulated Expression of a Diphtheria Toxin A-Chain Gene
Updated Version http://cancerres.aacrjournals.org/content/46/9/4660
Access the most recent version of this article at:
Citing Articles http://cancerres.aacrjournals.org/content/46/9/4660#related-urls
This article has been cited by 17 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and .pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions .permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
[CANCER RESEARCH 46, 4660-4664, September 1986)
Regulated Expression of a Diphtheria Toxin A-Chain Gene Transfected into
Human Cells: Possible Strategy for Inducing Cancer Cell Suicide1
Ian H. Maxwell,2 FrançoiseMaxwell, and L. Michael Glode
Division of Medical Oncology, University of Colorado Health Sciences Center, Denver, Colorado 80262
ABSTRACT
As an alternative to directing plant or bacterial toxins to surface
receptors, we are investigating the possibility of killing tumor cells by
the expression of an exogenously introduced toxin gene (i.e., cell suicide).
Tissue-specific gene regulatory elements might thus be exploited to
achieve selective killing. To assess the feasibility of such an approach,
we have transfected human cells (HeLa, B-lymphoblastoid, and 293 cells)
with plasmids containing the diphtheria toxin A-chain (DT-A) coding
sequence. The presence of the DT-A sequence lowered the level of
transient expression of chloramphenicol acetyltransferase from a cotrans-
fected plasmid, pSV2cat. This expression level in B-cells was further
diminished by the inclusion of an immunoglobulin enhancer in the DT-A
plasmid. In cotransfection experiments with a DT-A plasmid lacking an
enhancer, chloramphenicol acetyltransferase expression was much more
strongly inhibited in 293 cells (which express adenovirus EIA and E1B
products) than in the other cell types; furthermore, the presence of the
DT-A sequence eliminated recovery of G418-resistant 293 cell transform-
ants after transfection with a plasmid containing the neo selectable
marker. These results suggest that cell-specific regulatory mechanisms
can be exploited to achieve selective cell killing by expression of an
introduced toxin gene.
INTRODUCTION
Proteins which are highly toxic when introduced into mam
malian cells are produced by many species of plants and bacte
ria. For example, mouse L-cells are killed by the introduction
of DT-A3 at a concentration as low as one molecule/cell (1).
Attempts have been made to replace natural, B-chain-mediated
toxin entry (2) by mechanisms dependent on specific cell surface
molecules (e.g., hormone receptors or specific surface antigens)
with a view to obtaining selective lethality for certain tumor
cells. We are exploring an alternative means of exploiting a
natural toxin which does not depend on cell surface molecules.
Instead, DNA coding for DT-A is introduced into cells in
constructs containing tissue-specific, cis-acting, transcriptional
regulatory elements with the intention of achieving lethal
expression in cells containing the factors to which these ele
ments respond.
The production of enzymically active toxin in mammalian
cells by expression of the DT-A gene should result in the
inhibition of further protein synthesis (by ADP ribosylation of
elongation factor 2) (2) and consequent cell death. In this paper,
we present evidence that transfection with DT-A expression
plasmids produces toxic effects which can be modulated in a
cell-specific manner. By enabling the elimination of specific
target cell populations, this approach should find significant
applications in developmental and cell biology and eventually
in cancer therapy.
Received 2/14/86; revised 5/20/86; accepted 5/30/86.
The costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
1Supported by grants from Joyce Calvert, the Kyle Dudley Cancer Research
Foundation, and the Tad Beck Foundation; by Grant 84-37 from the Milheim
Foundation for Cancer Research, and by American Cancer Society Institutional
Grant IN-5Y.
1To whom requests for reprints should be addressed.
3The abbreviations used are: DT-A, diphtheria toxin A-chain; CAT, chlor
amphenicol acetyltransferase; MT, metallothionein.
MATERIALS AND METHODS
Plasmids and Cell Lines. DT-A plasmid, pi) 1201 (3), was supplied
by J. Murphy, pSV2/3-globin (4) by Paul Berg, p84H (containing MT
HA) (5) by M. Karin and A. Haslinger, pSER.Alu.a (6) by A. Hayday,
and pSV2cat (7) by B. Howard. 293 cells (8) were from J. Alwine and
the GM4025 B-lymphoblastoid line was from the Human Mutant Cell
Genetic Repository, Camden, NJ. HeLa and 293 cells were maintained
in Dulbecco's modified Eagle's medium with 10% newborn bovine
serum. GM4025 cells were maintained in RPMI 1640 medium with
10% fetal bovine serum. Media also contained gentamicin sulfate (SO
Mg/ml).
Plasmid Constructions (Fig. 1). A parental expression plasmid,
pXMT, was constructed by cloning part of the human metallothionein
IIA (MT IIA) promoter into a pSV2 (4) derivative as follows. pSV2-
327-/3G was constructed from pSV2/3-globin (4) by substituting a
pBR327 DNA fragment (Sall-Pstl) for pBR322 DNA (Pvull-Pstl), thus
removing the poison sequence (9) and introducing an Ym<;lll site. The
BamHl site in the SV40-derived DNA was mutated by partial BamHl
cleavage, filling in, and ligation. An Xmalll-Bglll MT IIA fragment
(—71to +272, relative to the cap site) (5) from plasmid p84H was
cloned into the corresponding sites of the above vehicle to give pXMT.
In the DT gene, a \i/«.V\Isite occurs at the boundary of the A and B
coding regions and a Hhal site is present in the first and second codons
of the mature A-chain (10, 11). pDT201 (3) consists of an 831-base
pair Sau3Al DNA fragment including the DT-A coding sequence,
cloned into pUC8. We cleaved pDT201 with Hhal, blunted the 3'-
overhang with T4 DNA polymerase and then, after Sau3M cleavage,
isolated the »580-base pair DT-A fragment. This was cloned into
pXMT, which had been cleaved at the unique l!am\\\ site (overlapping
the ATG at position +73 of MT IIA), filled in, and then cleaved with
Bglll (at the MT IIA-SV40 junction), to give pTH-1. The blunt end
ligation at the 5'-end joined the MT initiator ATG in-frame to the DT-
A coding sequence and regenerated the BamHl site. To obtain the
frame-shift mutant pTH-2, pTH-1 was cleaved at the unique Acci site
in the DT-A coding sequence (35 codons from the NH2 terminus), filled
in with Klenow DNA polymerase, and religated, resulting in a 2-base
pair insertion. Constructs pTH-3 and pTH-4, containing the 279-base
pair Alul immunoglobulin heavy chain enhancer fragment (6) in the
EcoRl site were made by recombining Pstl DNA fragments from pTH-
1 or pTH-2 and pSER.Alu.a (6). The neo transcriptional unit (contain
ing SV40 promoter, splicing, and polyadenylation signals), isolated
from a derivative of pSV2neo (4), was cloned into the EcoRI site of
pTH-1 or pTH-2 using linkers. pTH-lneo and pTH-leon differ only
in the orientation of the neo unit.
Plasmids were isolated using an alkaline sodium dodecyl sulfate lysis
procedure (12) which efficiently removed bacterial chromosomal DNA.
In most preparations, ribosomal RNA was then removed by precipita
tion with NaCl (~4 M at 0°Covernight) and, after phenol plus chloro
form extractions, plasmid was excluded from Sepharose CL2B. In some
preparations, plasmids were purified by isopyknic centrifugation in
CsCl plus ethidium bromide (13). Equivalent results were obtained in
transfections with the same plasmids purified by either procedure.
Transfections. HeLa and 293 cells were transfected (5 x 10s cells/
transfection) with plasmid DNA mixtures using calcium phosphate
coprecipitation (7, 14), including, for HeLa, a 20% v/v glycerol shock
(7). GM4025 cells were transfected (1 x IO7 cells/transfection) using
DEAE-dextran, followed by exposure to chloroquine (15, 16).
For assays of transient expression of CAT activity, cells were har
vested 48 h after transfection and extracts were prepared by freezing
and thawing 3 times. CAT activity was determined (7) by acetylation
of [l4C]chloramphenicol (0.1 nC\ at «45fÃ-Ci/^mol/150 n\ reaction)
4660
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
TOXIN GENE TRANSFECTION AND EXPRESSION
using samples containing 100-200 ¿igprotein (Bio-Rad assay). The
percentage of acetylation of the [l4C]chloramphenicol was determined
by scintillation counting of spots excised from the thin layer Chromat
ographie plate.
For selection of G418-resistant transformants, cells were trypsinized
2 days after transfection and replated in medium containing G418 (4)
(nominal concentration of active fraction, 400 ^g/ml). Macroscopic
colonies were counted after 17 days.
RESULTS
We constructed DT-A expression plasmids using a truncated
metallothionein promoter for DT-A transcription, to allow the
possibility of heavy metal-regulated expression. Similar trun
cation of the MT IIA promoter was reported to minimize basal
transcription while retaining metal response (5); however, as
described below, we obtained evidence that the basal transcrip
tion level from this promoter was sufficient for significant
expression of DT-A without exposure of transfected cells to
heavy metal ions.
Plasmid constructs are shown in Fig. 1. The mature DT-A
coding sequence from the plasmid pDT201 (3) was inserted
into an expression plasmid to give the construct pTH-1. Deriv
atives of pTH-1 were constructed (see Fig. 1) containing either
an immunoglobulin heavy chain enhancer (pTH-3, pTH-4), or
a neo transcription unit to permit selection with the antibiotic
G-418. Derivatives designated with even numbering (pTH-2,
pTH-4, pTH-2neo) contain a frameshift mutation in the DT-A
coding sequence.
* MT IIA »DT-A
pSV2ß-globin ¥ SV2-327-BG » pXMT » pTH~l
Promoter
(Hh)
XBAUlr>T
A(Bg)(B)
|
ATG
nTH-7 FRAMESHIFT
' (A)
AATAAA
pTH-3 ENHANCER I
DT-A
pTH-4 FRAMESHIFT
(A) ENHANCER [
DT-A J_
Fig. 1. DT-A expression plasmids. Plasmids with odd numbering (pTH-1,
pTH-3) contain the wild-type DT-A coding sequence; even numbering (pTH-2,
pTH-4) indicates a frame-shift mutation has been introduced into the DT-A. The
coding region for mature DT-A is joined to a 5'-truncated metallothionein
promoter DNA fragment (from the human MT IIA gene) (5) which also supplies
the ATG translation initiation codon. pTH-1 codes for a DT-A product differing
from authentic DT-A (193 amino acids) by 4 amino acid residues (2 at each
terminus), pilli and pTH-4 contain a 279-base pair Alul fragment from human
immunoglobulin heavy chain DNA shown to have enhancer activity in B-cells
(6). pTH-1 neo and pTH-2neo (not shown) contain the neo gene to permit selection
of transformants resistant to G418 (4). , pBR327 I )N.\: •.MT HA promoter
region: O, DT-A DNA; D, SV40 DNA. Positions of the initiation codon (ATG),
SV40 splice (A.), and polyadenylation signal (AATAAA), and the ampicillin
resistance gene (AMP') are indicated, as are relevant restriction sites (X, A'molll;
Hh, Hhai; B, BamW; A, Acci; Bg, Bglll; S, Sau3M; E, EcoRl). Sites destroyed
in the constructions are shown in parentheses.
We first sought evidence for a toxic effect of the transfected
DT-A gene using transient expression systems. Since routinely
available procedures result in transfection of a relatively small
fraction of cells, we used cotransfection with a plasmid contain
ing the cat gene (7). Expression of DT-A should result in
inhibition of protein synthesis and therefore of CAT expression
in cotransfected cells. Similar experiments have been used to
study activation (14, 17) or inhibition (14, 18) of transcription
from specific promoters by other fra/iî-actingproducts. As
shown in Fig. 2a and Table 1, the level of CAT expression in
HeLa cells two days after transfection was inhibited by 80-97%
when 5 ng pSV2cat (7) was cotransfected with an equal mass
of a DT-A-expressing plasmid (pTH-1, pTH-lneo, or pTH-3).
Concentration-dependent inhibition was observed with these
plasmids (Fig. 2a, tracks 3-5), inhibition being substantial with
as little as 0.3 ¿¿g(Table 1). In contrast, cotransfection with a
plasmid lacking the DT sequence (Fig. la, tracks 6 and 7), or
with the frame-shift mutants pTH-2 or pTH-4, did not inhibit
CAT expression from pSV2cat (Table 1). These mutants were
constructed by introducing a frame shift into the early part of
the DT-A coding sequence (see Fig. 1) to eliminate production
of active DT-A. Failure of these mutants to inhibit CAT expres
sion supports the conclusion that the inhibition observed with
the corresponding DT-A expressing plasmids was due to active
DT-A production, since the mutants differed in structure only
by a 2-base pair insertion.
To assess activity of the truncated promoter, a construct
analogous to pTH-1 (designated pCAT-1) was made in which
the cat gene replaced the DT-A coding sequence. Transfection
of this construct into HeLa cells indicated a significant basal
level of expression (0.5-3% of that from the SV40 early pro
moter, as determined in parallel transfections with pSV2cat)
(results not shown). Exposure of the cells to cadmium chloride
(2.5-5 MM)during the transient expression period stimulated
CAT expression 1.5- to 6-fold. As shown above, we observed
strong inhibition of CAT expression in cotransfection experi
ments with pTH-1 without exposure of the cells to heavy metal
ions; this inhibition was increased only slightly (1- to 2-fold) by
cadmium (not shown). The basal activity of the truncated MT
IIA promoter therefore appeared sufficient to generate inhibi
tory levels of DT-A. Although cadmium response was minimal,
we were able to demonstrate other means of regulation of DT-
A expression from the truncated MT IIA promoter in the
following experiments.
Several cellular genes are associated with enhancers (cis-
acting DNA sequences) which activate transcription in response
to tissue-specific frans-acting factors (19-22). We wished to
determine whether linkage with an enhancer could direct tissue-
specific expression of the DT-A gene; therefore, we transfected
a human B-lymphoblastoid cell line with pSV2cat together with
pTH-1 or pTH-2 or their derivatives into which a human Ig H
chain gene enhancer (6) had been cloned (pTH-3 and pTH-4;
see Fig. 1). Immunoglobulin enhancers are preferentially active
in B-cells and are among the best characterized cellular ele
ments regulating tissue-specific gene expression (6, 23, 24). To
confirm the effectiveness of the enhancer in the B-cell line we
made a plasmid derivative (pCAT-3) corresponding to pTH-3
in which the cat gene replaced the DT-A sequence (again using
the truncated MT promoter). CAT expression from pCAT-3 in
the B-cells was >25-fold that from pCAT-1 at 48 h posttrans-
fection (results not shown). In contrast, pCAT-1 and pCAT-3
gave approximately equal expression when transfected into
HeLa cells (not shown).
The effects of cotransfecting B-cells with pSV2cat and pTH-
4661
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
TOXIN GENE TRANSFECTION AND EXPRESSION
(a) HeLa
% ACETYLATED
83 91 33 19 7 83 80
(b)B-cells
51 43 30
3450.3
1pg pTH-lneo56
70.5
5ugpSV2-327neo
%ACETYLATED
24 8 22 9 0.8 33 39
Table 1 Transient CAT activity obtained after cotransfection ofpSV2cat with
plasmids containing wild-type or frame-shift mutant DT-A gene
Cells of each type were transfected as described in "Materials and Methods"
with 5 tig pSV2cat together with the indicated amounts of other plasmids. Results
are from multiple experiments using two independent preparations each of pTH-
1, -2, -3, and -4. Values are percentage of control CAT activity observed in the
same experiment with pSV2cat alone. Where >4 determinations were made, the
mean ±SD is shown.
HeLa cells, % of control CAT activitywithpSV2-327neo
pTH-2neo
pTH-2
pTH-4
pTH- 1neo
pTH-I
pTH-30.3
»g38
42 ±15 (8)°
56 ±23 (7)0.5«94
106,9023,46,
17
541
Pg65,
120
84
19
46± 12(11)
38 ±9(11)s««91137,
52
121, 179
89, 104,81
8, 7, 3, 3
10, 17, 15, 16
11,13,20B
cells, % of control CAT activitywithpTH-2
pTH-4
pTH-I
pTH-30.1
Mg65,
147
47,700.3
Mg148107
62 ±10(7)
33 ±13(7)1
Mg70,
105
83, 129
30 ±16 (7)
16 ±11 (6)3*ig69
56
15,21, 17
6,7,11293
cells, % of control CAT activitywithpTH-2
pTH-10.03
/ig180.1 Mg60.3 «97
5,3,3Ifg90 0.4, 1
" Numbers in parentheses, number of determinations.
•1•
•2
30.1pg•
•t•50.3
1
pTH-1•
60.1pg•7•8•
90.3
1 Ipg
pTH-3 pTH-2•
10Ipg
pTr
(c) 293 cells
ACETYLATED
28 2.1 1.7 32 30 27
®
0.03 0.1
pg pTH-1 0.3 0.3 1
pg pTH-2
Fig. 2. Inhibition of CAT expression from pSV2cat in (a) HeLa cells, (e) B-
lymphoblastoid cells, and (c) 293 cells by cotransfection with plasmids containing
the wild-type DT-A coding sequence. Cells were transfected with DNA mixtures
containing 5 «KpSV2cat and the indicated amounts of other plasmids or with 5
pg pSV2cat alone (tracks 1 and 2 in a and h and truck* 1 and 5 in c). The
autoradiogram of the thin layer chromatogram from the CAT assay is shown.
1, -2, -3, or -4 are shown in Fig. 2b and Table 1. The H-chain
enhancer increased inhibition of CAT expression such that ~3-
fold more pTH-1 than pTH-3 was required for the same level
of inhibition (Table 1; and compare Fig. 2b tracks 3-5 with 6-
8). Neither pTH-2 nor its derivative pTH-4 (DT-A frame-shift
control plasmids with or without enhancer) was inhibitory when
cotransfected in amounts up to 1 /¿g:the relatively minor
inhibition apparent with 3 /<gof these plasmids (Table 1) might
result from competition for cellular transcript ¡miaifactors (25,
26). In similar cotransfections of HeLa cells pTH-3 was no
more effective than pTH-1 in inhibiting CAT expression from
pSV2cat (Table 1). We conclude that the expression of a toxin
gene can be regulated in a cell-specific manner by linkage with
an enhancer. We are currently investigating whether preferen
tial DT-A expression in B-cells can be increased by substituting
an immunoglobulin promoter (27-29) for the metallothionein
promoter in pTH-3.
To explore further the possibility that trans-acting factors
modulating promoter or enhancer activity might serve to re
strict toxin gene expression to specific cell types, we performed
cotransfection experiments with 293 cells, a human embryonic
kidney cell line expressing the EIA and El B products of
adeno virus type 5 (8). The EIA product increases the transcrip-
tional activity of adenovirus early and certain cellular promoters
(14, 30) while repressing certain other promoters (14, 18). As
shown in Fig. 2c and Table 1, pTH-1 inhibited CAT expression
from cotransfected pSV2cat much more strongly in 293 than
in HeLa cells (Fig. 2a). As little as 30 ng pTH-1 inhibited CAT
expression from 5 ng pSV2cat by 80% (Fig. 2c, track 2), while
up to 1 ng of the frame-shift DT-A control plasmid pTH-2 did
not inhibit significantly (tracks 6 and 7). This suggests that the
greater inhibition observed in 293 cells resulted from increased
transcription from the MT HA promoter relative to the SV40
early promoter of pSV2cat so that the cells expressed a higher
ratio of DT-A to CAT enzyme. These effects may involve
differential activation/repression of the 2 promoters by the EIA
4662
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
TOXIN GENE TRANSFECTION AND EXPRESSION
Table 2 Frequency ofC4I8 resistant transformants obtained after transfection of
293 cells with neo plasmids containing wild-type orframe-shift mutant DT-A
gene
293 cells were transfected with plasmids (10 pg) containing the neo transcrip
tion unit. Plasmids used (see Fig. 1) were: controls, pSV2-327neo (no DT-A) and
pTH-2neo (frame-shift mutant DT-A); with wild-type DT-A, pTH-1 neo and pTH-
leon (pTH-1 derivatives differing only in the orientation of the neo transcription
unit).
PlasmidNone
pSV2-327neo
pTH-lneo
pTH-leon
pTH-2neoNumber
ofcolonies064
0,0
0
79,71
product (14, 18), in addition to the unusual stabilization of
transfected plasmid DNA observed in 293 cells (14).
To obtain additional evidence for toxicity resulting from
expression of the DT-A gene, we transfected 293 cells with neo-
containing derivatives of pTH-1 or pTH-2 and selected for
transformants resistant to G418 (4). As shown in Table 2, no
transformants were recovered after transfection with the DT-
A-expressing plasmids pTH-lneo and pTH-leon; in contrast,
the DT-A frame-shift mutant pTH-2neo gave a similar trans
formation frequency («1.4x 10~4)to that observed with a neo
plasmid containing no DT-A sequence. We infer that failure to
recover transformants from the pTH-1 derivatives resulted from
a lethal level of expression of DT-A in the transfected cells;
however, in similar experiments with HeLa cells (results not
shown), transformants were obtained with pTH-lneo with an
average frequency approximately one-half of that found with
pTH-2neo (which was ~1 x 10~5);thus, a minimal level of DT-
A expression such as that obtained by transcription from the
truncated MT HA promoter in HeLa may not necessarily be
incompatible with cell survival. This conclusion suggests the
feasibility of protecting non-target cells from toxicity.
DISCUSSION
Our findings of inhibition of gene expression from a cotrans-
fected plasmid and lowered transformation frequency are con
sistent with the conclusion that transfection of an efficiently
expressed DT-A gene is lethal. We have demonstrated two ways
in which the expression of a transfected DT-A gene can be
modulated in different cell types: (a) expression was increased
in B-cells but not in HeLa cells when an immunoglobulin
enhancer was present in the construct; (b) expression from a
weak promoter, lacking known enhancer elements, was much
stronger in 293 cells than in HeLa or B-cells. We have so far
demonstrated preferential rather than specific expression of
DT-A in different cell types. Rapidly increasing knowledge of
tissue-specific gene control mechanisms will be directly appli
cable to designing efficient means of directing the expression
of exogenous genes including DT-A to specific cell types. At
tenuated DT-A mutants are available (1, 2), should control of
basal expression of the wild-type gene prove inadequate to avoid
toxicity in non-target cells. The achievement of tightly con
trolled tissue-specific lethality following introduction of a toxin
gene into cells is therefore a realistic near-term goal.
With regard to the eventual application of toxin gene therapy
to cancer, our results indicate that immunoglobulin enhancers
might be used to contribute to target specificity in treatment of
B-cell neoplasms. Although normal B-cells would also be sus
ceptible, the toxicity to other dividing cell populations seen
with current therapeutic agents would be avoided. Tissue-spe
cific enhancers from other genes coding for "tumor-specific"
products might similarly be harnessed in directing expression
of a toxin gene specifically in tumor cells. Promising candidates
would include genes coding for products normally expressed
only in fetal tissues but frequently expressed ectopically in
certain tumors (31). The enhanced DT-A expression we ob
served in 293 cells probably resulted at least partly from the
presence of adenovirus EIA products. It may therefore also be
possible to exploit the abundant expression in certain tumors
(32) of proteins functionally analogous to EIA (e.g., the c-myc
product) (33, 34) to achieve selective killing by a toxin gene.
These concepts potentially offer a novel approach to cancer
therapy less subject to undesirable side effects than current
procedures. It is clear that the success of such therapy will
depend on the development of efficient means of delivering
exogenous genes to tumor cells both in terms of access to and
efficient uptake by these cells. Intensive work on recombinant
viruses has produced vehicles capable of efficient transduction
of foreign genes into cells In vitro (35-38). Such defective
recombinant viruses should be applicable to the delivery of
toxin gene therapy to accessible populations of cancer cells,
e.g., in bone marrow, or in skin or bladder cancers. Although
systemic delivery of toxin gene vectors to solid tumors or
métastaseswill be more difficult, this problem may also even
tually be overcome using viral vehicles by exploiting the mech
anisms evolved by many viruses for efficiently infecting tissues
remote from their site of entry into the organism.
ACKNOWLEDGMENTS
We thank Paula Lane for skillful assistance.
REFERENCES
1. Yamaizumi, M., Mekada, E., Uchida, T., and Okada, Y. One molecule of
diphtheria toxin fragment A introduced into a cell can kill the cell. Cell, 15:
245-250, 1978.
2. Pappenheimer, A. M., Jr. Diphtheria toxin. Ann. Rev. Biochem., 46:69-94,
1977.
3. Leong, I>., Coleman, K. I)., and Murphy, J. R. Cloned fragment A of
diphtheria toxin is expressed and secreted into the periplasmic space of
Escherichia coli K12. Science (Wash. DC), 220: 515-517, 1983.
4. Southern, P. J., and Berg, P. Transformation of mammalian cells to antibiotic
resistance with a bacterial gene under control of the SV40 early region
promoter. J. Mol. Appi. Genet., /.- 327-341, 1982.
5. Karin, M., Haslinger, A., Holtgreve, H., Richards, R. I., Krauter, P., West-
•phal,H. M., and Beato, M. Characterization of DNA sequences through
which cadmium and glucocorticoid hormones induce human metallothionein-
HAgene. Nature (Lond.), 308: 513-519, 1984.
6. Hayday, A. C, Gillies, S. p., Saito, H., Wood, C., Wiman, K., Hayward, W.
S., and Tonegawa, S. Activation of a translocated human c-myc gene by an
enhancer in the immunoglobulin heavy-chain locus. Nature (Lond.), 307:
334-340, 1984.
7. Gorman, C. M., Moffat, L. F., and Howard, B. H. Recombinant genomes
which express chloramphenicol acetyltransferase in mammalian cells. Mol.
Cell. Biol., 2:1044-1051, 1982.
8. Graham, F. L., Smiley, J., Russell, W. C., and Nairn, R. Characteristics of a
human cell line transformed by DNA from human adenovirus type 5. J. Gen.
Viro!., 56:59-72, 1977.
9. Lusky, M., and Botchan, M. Inhibition of SV40 replication in simian cells
by specific pBR322 DNA sequences. Nature (Lond.), 293: 79-81, 1981.
10. Greenfield, L., Björn,M. J., Horn, G., Fong, D., Buck, G. A., Collier, R. J.,
and Kaplan, D. A. Nucleotide sequence of the structural gene for diphtheria
toxin carried by corynebacteriophage ß.Proc. Nati. Acad. Sci. USA, 80:
6853-6857, 1983.
11. Kaczorek, M., Delpeyroux, F., Chenciner, N., Streeck, R. E., Murphy, J. R.,
Boquet, P., and Tiollais, P. Nucleotide sequence and expression of the
diphtheria w.v228 gene in Escherichia coli. Science, 227:855-858, 1983.
12. Ish-Horowicz, D., and Burke, J. F. Rapid and efficient cosmici cloning.
Nucleic Acids Res., 9: 2989-2998, 1981.
13. T. Maniatis, E. F. Fritsch, and J. Sambrook (eds.). Molecular Cloning, p.
93. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 1982.
14. Alwine, J. C. Transient gene expression control: effects of transfected DNA
stability and trans-activation by viral early proteins. Mol. Cell. Biol., 5:1034-
1042, 1985.
15. McCutchan, J. H., and Pagano, J. S. Enhancement of the infectivity of
4663
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
TOXIN GENE TRANSFECTION AND EXPRESSION
simian virus 40 deoxyribonucleic acid with diethylaminoethyl-dextran. J.
Nail. Cancer Inst., 41: 351-357, 1968.
16. Luthman, II., and Magnusson, G. High efficiency polyoma DNA transfection
of chloroquine treated cells. Nucleic Acids Res., 11: 1295-1308, 1983.
17. Kingston, R. E., Baldwin, A. S., Jr., and Sharp, P. A. Regulation of heat
shock protein 70 gene expression by c-myc. Nature (Lond.), 312: 280-282,
1984.
18. Velcich, A., and Ziff, E. Adenovirus Eia proteins repress transcription from
the SV40 early promoter. Cell, 40: 705-716, 1985.
19. Gillies, S. D., Folsom, V., and Tonegawa, S. Cell type-specific enhancer
element associated with a mouse MHC gene, Eß.Nature (Lond.), 310: 594-
597, 1984.
20. Walker, M. D., Edlund, T., Boulet, A. M., and Rutter, W. J. Cell-specific
expression controlled by the 5'-flanking region of insulin and chymotrypsin
genes. Nature (Lond.), 306: 557-561, 1983.
21. Ornitz, D. M., Palmiter, R. D., Hammer, R. E., Brinster, R. L., Swift, G.
II., and MacDonald, R. J. Specific expression of an elastase-human growth
hormone fusion gene in pancreatic acinar cells of transgenic mice. Nature
(Lond.), 5/5:600-602,1985.
22. Ephrussi, A., Church, G. M., Tonegawa, S., and Gilbert W. B-Lineage-
specific interactions of an immunoglobulin enhancer with cellular factors in
vivo. Science (Wash. DC), 227: 134-140, 1985.
23. Gillies, S. D., Morrison, S. L., Oi, V. T., and Tonegawa, S. A tissue-specific
transcription enhancer element is located in the major intron of a rearranged
immunoglobulin heavy chain gene. Cell, 33: 717-728, 1983.
24. Picard, D., and Schaffner, W. A lymphocyte-specific enhancer in the mouse
immunoglobulin k gene. Nature (Lond.), 307: 80-82, 1984.
25. Mercóla, M., Goverman, J., Mirell, C., and CaÃ-ame,K. Immunoglobulin
heavy-chain enhancer requires one or more tissue-specific factors. Science
(Wash. DC), 227: 266-270, 1985.
26. Seguin, C., Felber, B. K., Carter, A. D., and Hamer, D. H. Competition for
cellular factors that activate metallothionein gene transcription. Nature
(Lond.), 312: 781-785, 1984.
27. Foster, J., Stafford, J., and Queen, C. An immunoglobulin promoter displays
cell-type specificity independently of the enhancer. Nature (Lond.), 315:423-
425, 1985.
28. Mason, J. O., Williams, G. T., and Neuberger, M. S. Transcription cell type
specificity is conferred by an immunoglobulin v,, gene promoter that includes
a functional consensus sequence. Cell, 41:479-487, 1985.
29. Grosschedl, R., and Baltimore, D. Cell-type specificity of immunoglobulin
gene expression is regulated by at least three DNA sequence elements. Cell,
47:885-897, 1985.
30. Stein, R., and Ziff, E. B. HeLa cell /3-tubulin gene transcription is stimulated
by adenovirus 5 in parallel with viral early genes by an E1a-dependent
mechanism. Mol. Cell. Biol., 4: 2792-2801, 1984.
31. Mclntire, K. R. Tumor markers: how useful are they? Hosp. Pract., 19: 55-
68, December 1984.
32. Little, C. D., Nau, M. M., Carney, D. N., Gazdar, A. F., and Minna, J. D.
Amplification and expression of the e-myc oncogene in human lung cancer
cell lines. Nature (Lond.), 306: 194-196,1983.
33. Land, H., Parada, L. F., and Weinberg, R. A. Tumorigenic conversion of
primary embryo fibroblasts requires at least two cooperating oncogenes.
Nature (Lond.), 304: 596-602, 1983.
34. Ruley, H. E. Adenovirus early region 1A enables viral and cellular transform
ing genes to transform primary cells in culture. Nature (Lond.), 304: 602-
606, 1983.
35. Tratschin, J.-D., West, M. H. P., Sandbank, T., and Carter, B. J. A human
parvovirus, adeno-associated virus, as a eukaryotic vector: transient expres
sion and encapsidation of the prokaryotic gene for chloramphenicol acetyl-
transferase. Mol. Cell. Biol., 4: 2072-2081, 1984.
36. Karlsson, S., Humphries, R. K., Gluzman, Y., and Nienhuis, A. W. Transfer
of genes into hematopoietic cells using recombinant DNA viruses. Proc.
Nati. Acad. Sci. USA, 82: 158-162, 1985.
37. Miller, A. D., Law, M.-F., and Verma, I. M. Generation of helper-free
amphotropic retroviruses that transduce a dominant-acting, methotrexate-
resistant dihydrofolate reducÃ-asegene. Mol. Cell. Biol., 5:431-437, 1985.
38. Cone, R. D., and Mulligan, R. C. High-efficiency gene transfer into mam
malian cells: generation of helper-free recombinant retrovirus with broad
mammalian host range. Proc. Nati. Acad. Sci. USA, 81: 6349-6353, 1984.
4664
American Association for Cancer Research Copyright © 1986 on July 10, 2011cancerres.aacrjournals.orgDownloaded from
... Its action requires the expression of DTR on living cells (5). Following cell surface binding of DT-B and receptormediated endocytosis of DTR, subunit A (DT-A, or DTA) translocates from the acidic late endosome to the cytosol where it ADP-ribosylates a diphthamide residue of elongation factor 2 (6, 7), terminating protein synthesis (8)(9)(10)(11) and ultimately triggering cell death (12). Although DT-A acts on all eukaryotic elongation factor 2, murine and rat cells are insensitive to DT (13,14). ...
Article
Full-text available
Genetic technology using site-specific recombinases (SSR), such as the Cre-loxP system, has been widely employed for labelling specific cell populations and for studying their functions in vivo. To enhance the precision of cell lineage tracing and functional study, a similar SSR system termed Dre-rox has been recently used in combination with Cre-loxP. To enable more specific cell lineage tracing and ablation through dual recombinase activity, we generated two mouse lines that render Dre- or Dre+Cre-mediated recombination to excise a stop codon sequence that prevents the expression of diphtheria toxin receptor (DTR) knocked into the ubiquitously expressed and safe Rosa26 locus. Using different Dre- and Cre-expressing mouse lines, we showed that the surrogate gene reporter tdTomato and DTR were simultaneously expressed in target cells and in their descendants, and observed efficient ablation of tdTomato⁺ cells after diphtheria toxin administration. These mouse lines were used to simultaneously trace and deplete target cells of interest through the inducible expression of a reporter and DTR using dual Cre and Dre recombinases, allowing more precise and efficient study of the role of specific cell subsets within a heterogeneous population in pathophysiological conditions in vivo.
... First, we utilized a genetic approach to selectively ablate TRPV1-expressing nociceptors by crossing TRPV1-Cre mice that expressed Cre recombinase under the control of TRPV1 locus, with floxed diphtheria toxin A (DTA) mice (Chiu et al., 2013). TRPV1-driven expression of DTA induces cell death by catalyzing the inactivation of elongation factor 2 and inhibiting protein synthesis (Collier, 2001;Maxwell et al., 1986;Palmiter et al., 1987). We have previously established that TRPV1-Cre/DTA mice do not express TRPV1+ neurons (Zanos et al., 2018). ...
Preprint
Full-text available
Mammals store memories in the nervous and immune systems. Sensory neurons have been implicated in enhancing neurological memory, but whether neurons participate during immunity to novel antigens is unknown. Here, mice rendered deficient in transient receptor potential vanilloid 1 (TRPV1)-expressing sensory neurons, termed “nociceptors,” fail to develop competent antibody responses to KLH and hapten-NP. Moreover, selective optogenetic stimulation of TRPV1 neurons during immunization significantly enhanced antibody responses to antigens. Thus, TRPV1 nociceptors mediate antibody responses to novel antigen, and stimulating TRPV1 nociceptors enhances antibody responses during immunization. This is the first genetic and selective functional evidence that nociceptors are required during immunization to produce antigen-specific antibodies. Summary The first genetic and selective functional evidence showing that TRPV1-expressing nociceptors are required for competent antibody responses to novel antigen, and stimulating TRPV1 nociceptors enhances antibody responses to novel antigen.
... However, to avoid unintended side effects on normal cells, this potent toxin requires efficient and reliable selective targeting. Several attempts have been done to target the DT-A toxicity, e.g. by modifying the promoter [188] or replacing the wild type DT-A sequence with attenuated mutant variants [189], with dissatisfying outcome. (2) DT binds to its receptor (Heparin-binding epidermal growth factor precursor). (3) Furin protease cleaves the polypeptide chain between C and T domain. ...
Thesis
Full-text available
Bakterielle Toxine stellen eine wirkungsvolle und effektive Alternative zur Therapie von Tumorerkrankungen dar. Das vom Clostridium perfringens Typ A produzierte Clostridium perfringens enterotoxin (CPE) gehört zu der Gruppe der porenbildenden Toxine und weist eine rezeptorspezifische zytotoxische Wirkung auf, welche über die Membranrezeptoren Cldn3 und Cldn4 entfaltet wird. Diese liegen vor allem in Epithelialkarzinomen wie dem Brust-, Prostata-, oder Kolon-, sowie dem Pankreaskarzinom (PK) stark hochreguliert vor. Ziel dieser Arbeit war die Anwendung des neuen selektiven und effizienten „Onkoleaking“ Suizid-Gentherapie Konzepts für die Behandlung von Cldn3 / 4 überexprimierender PK unter Verwendung eines nicht-viralen translations-optimierten CPE exprimierenden Vektors (optCPE). Weiterhin sollte in dieser Arbeit der genaue molekulare Mechanismus der CPE-vermittelten Zytotoxizität in vitro und auch in vivo analysiert werden. Für die in vitro Analysen wurden verschiedene humane PK Zelllinien, Patienten abgeleitete Xenotransplantate (PDX) und deren abgeleiteten Zellen bezüglich ihrer Cldn3 / 4 Expression und Sensitivität sowohl gegenüber rekombinantem CPE (rekCPE) als auch nach optCPE Gentransfer untersucht. Es konnte eine positive Korrelation zwischen der Effizienz CPE vermittelter Zytotoxizität und der Höhe der Cldn3 / 4 Überexpression gezeigt werden. Des Weiteren wurde die Verfügbarkeit und Zugänglichkeit der CPE Rezeptoren für die Toxinbindung als kritischer Faktor für die durch Porenbildung induzierte Zytotoxizität beschrieben. Auch eine detaillierte Analyse verschiedener apoptotischer und nekrotischer Signalwege und deren Schlüsselmoleküle waren vom besonderen Interesse. Von noch größerer Wichtigkeit war jedoch die Anwendbarkeit und der Nachweis der antitumoralen Wirksamkeit der optCPE-basierten Suizid-Gentherapie mit Hilfe des intratumoralen Jet-Injektion Gentransfers in verschiedenen Luziferase-exprimierenden CDX und PDX Modellen des PK. Alle in vivo Studien zeigten eine selektive optCPE vermittelte Verminderung der Tumorvitalität in Verbindung mit Nekrose, die in fast allen Fällen mit einer Reduktion des Tumorvolumens einher ging. Die tierexperimentellen Studien belegen damit die Effektivität der CPE-basierten Gentherapie im Pankreaskarzinom. Mit diesen neu gewonnenen Erkenntnissen zum „Onkoleaking“ Konzept der CPE Suizid-Gentherapie und deren Wirkungsmechanismen sind Kombinationen mit konventionellen Therapien möglich.
... In particular, interactions between microglia and NPCs regulate neurogenesis via phagocytosis and secretion of cytokines and chemokines [104][105][106][107]. Astrocytes in turn induce neuronal differentiation of NPCs via the release of neurogenic factors [108][109][110]. For example, it has been shown that astrocytes produce brain-derived neurotrophic factor (BDNF) neurotrophins that regulate hippocampal neurogenesis [111][112][113][114]. Neurogenesis is regulated via several mechanisms and at different levels, including the network and local circuit level [115,116], neuromodulatory level like serotonin (5-HT), norepinephrine (NE), dopamine (DA), and acetylcholine (ACh) [117][118][119][120], local signaling level like astrocytes [110], and other extrinsic factors level like exercise, stress and diet [102,109,[121][122][123][124]. Recently, 3D neurovascular tissues were constructed by combining in vitro neurogenesis and angiogenesis models using a microfluidic platform [125,126], where a triculture of human NSCs, human brain microvascular ECs (BMECs) and human mesenchymal stem cells (MSCs) was combined [125]. ...
Article
Full-text available
Understanding the mechanisms that govern nervous tissues function remains a challenge. In vitro two-dimensional (2D) cell culture systems provide a simplistic platform to evaluate systematic investigations but often result in unreliable responses that cannot be translated to pathophysiological settings. Recently, microplatforms have emerged to provide a better approximation of the in vivo scenario with better control over the microenvironment, stimuli and structure. Advances in biomaterials enable the construction of three-dimensional (3D) scaffolds, which combined with microfabrication, allow enhanced biomimicry through precise control of the architecture, cell positioning, fluid flows and electrochemical stimuli. This manuscript reviews, compares and contrasts advances in nervous tissues-on-a-chip models and their applications in neural physiology and disease. Microplatforms used for neuro-glia interactions, neuromuscular junctions (NMJs), blood-brain barrier (BBB) and studies on brain cancer, metastasis and neurodegenerative diseases are addressed. Finally, we highlight challenges that can be addressed with interdisciplinary efforts to achieve a higher degree of biomimicry. Nervous tissue microplatforms provide a powerful tool that is destined to provide a better understanding of neural health and disease.
Chapter
Gene therapy has started in the late 1980s as novel, clinically applicable therapeutic option. It revolutionized the treatment of genetic diseases with the initial intent to repair or replace defective genes. Gene therapy has been adapted for treatment of malignant diseases to improve the outcome of cancer patients. In fact, cancer gene therapy has rapidly gained great interest and evolved into a research field with highest proportion of research activities in gene therapy. In this context, cancer gene therapy has long entered translation into clinical trials and therefore more than two-thirds of all gene therapy trials worldwide are aiming at the treatment of cancer disease using different therapeutic strategies. During the decades in cancer gene therapy, tremendous knowledge has accumulated. This led to significant improvements in vector design, transgene repertoire, more targeted interventions, use of novel gene therapeutic technologies such as CRISPR/Cas, sleeping beauty vectors, and development of effective cancer immunogene therapies. In this chapter, a brief overview of current key developments in cancer gene therapy is provided to gain insights into the recent directions in research as well as in clinical application of cancer gene therapy.
Article
DNA including the coding sequence for the A chain of the mutant diphtheria toxin tox 176 was cloned. The cloned mature A-chain coding sequence showed a G-to-A transition at nucleotide 383 as the only difference from the wild-type sequence. This resulted in replacement of the glycine at position 128 by aspartic acid in the predicted amino acid sequence. A eucaryotic cell expression plasmid, pTH1-176, was constructed in which the tox 176 A-chain coding sequence was attached to a truncated metallothionein promoter. The toxicity of this construct, compared with that of the corresponding wild-type diphtheria toxin A-chain plasmid, pTH1, was assessed after transfection into the human 293 cell line by an indirect transient expression assay (I. H. Maxwell, F. Maxwell, and L. M. Glode, Cancer Res. 46:4660-4664, 1986). For the same effect, 15- to 30-fold more pTH1-176 than pTH1 was required, a result consistent with previous in vitro estimates of the diminished activity of the tox 176 A chain. Controlled expression of the cloned tox 176 A-chain coding sequence may provide a means of eliminating specific cell populations in an organism, for which purpose the wild-type diphtheria toxin A chain might prove too toxic.
Chapter
Genetic mouse models facilitate investigation of mechanisms underpinning human diseases and aid the development of novel therapeutic treatments. To better understand the demyelination and remyelination processes in adult-onset demyelinating diseases like multiple sclerosis (MS), we have developed the DTA mouse model system that allows for the widespread ablation of the mature oligodendrocytes, resulting in demyelination throughout the central nervous system (CNS). Induction of oligodendrocyte death in young adult DTA mice causes extensive CNS demyelination that leads to a severe neurological disease, followed by a full recovery that is associated with extensive replenishment of oligodendrocytes and remyelination. Thus, the DTA mouse enables investigation of the mechanisms that promote remyelination in MS and other adult-onset demyelinating diseases. Approximately 30 weeks later, the recovered DTA mice develop a fatal secondary demyelinating disease that is mediated by autoimmune T cells. Therefore, the DTA mouse model is also ideal for elucidating the role of oligodendrocyte death in eliciting autoimmunity in MS. In this chapter we describe the methods we used to generate the DTA mouse model and to analyze both the primary and secondary demyelinating diseases in DTA mice.
Chapter
The continuous integration of young neurons into the adult brain represents a novel form of structural plasticity and has inspired the creation of numerous computational models to understand the functional role of adult neurogenesis. These computational models consist of abstract models that focus on the utility of new neurons in simple neural networks and biologically based models constrained by anatomical data that explore the role of new neurons in specific neural circuits such as the hippocampus. Simulation results from both classes of models have suggested a number of theoretical roles for neurogenesis such as increasing the capacity to learn novel information, promoting temporal context encoding, and influencing pattern separation. In this review, we discuss strategies and findings of past computational modeling efforts, current challenges and limitations, and new computational approaches pertinent to modeling adult neurogenesis.
Chapter
Cancer is a disease of multifactorial etiology. Chemical, physical and viral environmental factors are involved in carcinogenesis. Numerous mutagenic events are required to transform a normal cell into a malignant one (1, 2). There is a high correlation between cancer and traumatic psychosocial events (3, 4). Carcinogenesis is a consequence of imbalances in the regulatory mechanisms that normally control the gene expression or function of the growth factors, receptors, and pathways of signal transduction. Chemical and physical carcinogens may induce cancer by altering the structure and functions of cellular protooncogenes. The viruses, however, transform cells either directly through an active viral oncogene or, if they lack oncogene, they may transform the cell by insertion of viral promoter sequences near or within the protooncogenes that would cause activation of protooncogene transcription and expression (2, 5-17). Discovery of oncogenes and their expression, as well as identification of their products, has thus revolutionized our approach to the molecular and genetic basis of multistage carcinogenesis. Protooncogenes normally code for proteins that play a role in the regulation of gene expression and at various stages of the action of growth factors during development and differentiation. These cellular protooncogenes do not seem to be tumorigenic themselves but must rather be activated by mutations and/or aberrant signal transduction and abnormal gene expression (1-17). The genes that code for receptors or components of signal transducing mechanisms, as well as for steroids, prolactin prostaglandins and other growth factors, should be considered as protooncogenes too, since the alteration of their functioning may contribute to abnormal growth and tumorigenesis (2,4,5,14,15). Malignant cells are immortalized, live longer than normal cells and fail to undergo normal differentiation. Their growth is autonomous, self-stimulated by autocrine secretion of various growth factors (2, 5-7). Immortalization seems to be associated with increased transcription of myc, myb, fos, jun oncogenes (2,5,6,8-11,13-17) and with the large T-gene of polyoma virus (5, 12).
Article
We present a procedure for cosmid cloning that allows rapid and efficient cloning of individual DNA fragments of between 32kb and 45kb. By appropriate treatment of the cloning vector, pJb8, we make left-hand and right-hand vector ends that are incapable of self-ligation but which accept dephosporylated insert DNA fragments. The inserted fragments are generated by partial digestion with MboI or Sau3A and are dephosphorylated to prevent ligation and insertion of non-contiguous fragments. The method eliminates the need to size the insert DNA fragments and prevents formation of clones containing short or multiple inserts. 1 microgram of target Drosophila DNA gives about 5 x 10(5) clones, with an average insert size of 38kb. We also describe a rapid and efficient method for preparing plasmid and cosmid DNA.
Article
Class II molecules of the major histocompatibility complex (MHC) are heterodimeric glycoproteins expressed on the surface of antigen-presenting B lymphocytes and macrophages1. The genes encoding the alpha- and beta-chains of the class II heterodimers, AalphaAbeta and EalphaEbeta, have recently been characterized at the molecular level2-5, and certain cloned genes were shown to be functionally expressed after introduction into cells by DNA-mediated gene transfer6,7. One study7 found that a transfected Ebbeta gene was expressed in a macrophage cell only after treatment of cells with gamma-interferon. DNA sequences associated with transfected Class II MHC genes may therefore have a regulatory role in their cell type-specific expression. We report here the identification of a cell type-specific transcriptional enhancer element associated with the mouse Edbeta gene.
Article
Erythrocyte ghosts containing a known number of molecules of purified fragment A of diphtheria toxin with a constant amount of FITC-BSA as a fluorescence marker were prepared by dialyzing a mixture of erythrocytes and these substances against hypotonic solution. These substances were then introduced into diphtheria toxin-resistant mouse L cells by virus-mediated cell fusion of the cells with the ghosts, and mononuclear recipients that has fused with only one erythrocyte ghost were separated in a flourescence-activated cell sorter (FACS) on the basis of their cell size and fluorescence intensity. After separation, the viability of cells containing known numbers of fragment A was examined by measuring colony-forming ability. The results demonstrated that a single molecule of fragment A was sufficient to kill a cell. This fact was confirmed by introduction into cells of fragment A from an immunologically related mutant toxin, CRM 176 (fragment A176); this has a completely functional fragment B region, but in cell extracts, the enzymic activity of its fragment A is about 10 fold less than that of wild toxin. The cytotoxicity of CRM 176 is about two hundredths of that of the wild-type (Uchida, Pappenheimer and Greany, 1973). As expected, about 100-200 fold excess of fragment A-176 was needed to kill the cells.
Article
The ability of recombinant DNA viruses to transfer genes into hematopoietic cells has been explored. A recombinant simian virus 40 (SV40) in which the early region had been replaced with the chloramphenicol acetyltransferase (CAT) gene driven by the promoter from Rous sarcoma virus (RSV), was constructed. This virus transferred the CAT gene more efficiently into mouse and human bone marrow cells and into the K562, MEL, and WEHI hematopoietic tissue culture cell lines, than the classical calcium phosphate DNA transfer procedure, as shown by assay for CAT activity 48 hr after infection. Recombinant SV40 virions were also shown to be capable of stably transforming Chinese hamster ovary cells by use of an early region recombinant containing the methotrexate-resistant dihydrofolate reductase (DHFR) gene driven by the RSV promoter. The entire DHFR transcriptional unit could be detected in the genome of transformed cells that were also shown to be resistant to methotrexate. A recombinant adenovirus stock containing the neomycin-resistance gene driven by the SV40 early promoter was used to infect the K562 and MEL hematopoietic cell lines to resistance to the antibiotic G418. Transformation frequency was 10- to 100-fold higher than that obtained with calcium phosphate-precipitated DNA. Most or all of the recombinant adenovirus genome was integrated as 1-3 copies in the transformed cells. These studies show the feasibility of using DNA viruses for introduction of new genetic material into hematopoietic cells.
Article
Using a transient expression assay in HeLa cells, we show that products from the adenovirus-5 E1a transcription unit repress transcription from the SV40 early promoter. The repression is unrelated to T antigen autoregulation, occurs maximally with low concentrations of E1a expression plasmid, is exerted at the transcriptional level, and requires functional E1a protein. The 289 and 243 amino acid E1a proteins are equally effective at repressing transcription. Since only the 289 amino acid protein is efficient at activating transcription, we conclude that activation and repression are separate E1a functions. We discuss possible mechanisms for E1a repression and the relationship of repression to the function of E1a in cell immortalization and transformation.
Article
We constructed several retroviruses which transduced a mutant dihydrofolate reductase gene that was resistant to methotrexate inhibition and functioned as a dominant selectable marker. The titer of dihydrofolate reductase-transducing virus produced by virus-producing cells could be increased to very high levels by selection of the cells in increasing concentrations of methotrexate. Helper virus-free dihydrofolate reductase-transducing virus was also generated by using a broad-host-range amphotropic retroviral packaging system. Cell lines producing helper-free dihydrofolate reductase-transducing virus with a titer of 4 X 10(6) per ml were generated. These retroviral vectors should have general utility for high-efficiency transduction of genes in cultured cells and in animals.
Article
The effects of trans-acting factors and transfected DNA stability on promoter activity were examined with chloramphenicol acetyl transferase (CAT) transient expression analysis. With cotransfection into CV-1P and HeLa cells, simian virus 40 T antigen, adenovirus E1a, and herpes-virus IE proteins were compared for their ability to trans-activate a variety of eucaryotic promoters constructed into CAT plasmids. T antigen and the IE protein were promiscuous activators of all the promoters tested [the simian virus 40 late promoter, the adenovirus E3 promoter, the alpha 2(I) collagen promoter, and the promoter of the Rous sarcoma virus long terminal repeat]. Conversely the E1a protein was specific, activating only the adenovirus E3 promoter and suppressing the basal activity of the other promoters. This specificity of activation by E1a contrasted with the high activity generated by all of the promoter-CAT plasmids when transfected into 293 cells, which endogenously produce E1a protein. Examination of transfected 293 cells determined that they stabilized much greater amounts of plasmid DNA than any other cells tested (CV-1P, COS, NIH-3T3, KB). Thus the high activity of nonadenovirus promoter-CAT plasmids in 293 cells results from the cumulative effect of basal promoter activity from a very large number of gene copies, not from E1a activation. This conclusion was supported by similar transfection analysis of KB cell lines which endogenously produce E1a protein. These cells stabilize plasmid DNA at a level comparable to that of CV-1P cells and, in agreement with the CV-1P cotransfection results, did not activate a nonadenovirus promoter-CAT plasmid. These results indicate that the stability of plasmid DNA must be considered when transient gene expression is being compared between cell lines. The use of relative plasmid copy numbers for the standardization of transient expression results is discussed.