You are on page 1of 6

GENE THERAPY

Correction of sickle cell disease by homologous recombination


in embryonic stem cells
Li-Chen Wu, Chiao-Wang Sun, Thomas M. Ryan, Kevin M. Pawlik, Jinxiang Ren, and Tim M. Townes

Previous studies have demonstrated that copy of the gene (␤A). We recently derived These experiments provide a foundation
sickle cell disease (SCD) can be cor- embryonic stem (ES) cells from a novel for similar studies in human ES cells
rected in mouse models by transduction knock-in mouse model of SCD and tested derived from sickle cell patients. Al-
of hematopoietic stem cells with lentiviral a protocol for correcting the sickle muta- though efficient methods for production
vectors containing antisickling globin tion by homologous recombination. In of human ES cells by somatic nuclear
genes followed by transplantation of these this paper, we demonstrate the replace- transfer must be developed, the data in
cells into syngeneic recipients. Although ment of the human ␤S-globin gene with a this paper demonstrate that sickle cell
self-inactivating (SIN) lentiviral vectors human ␤A-globin gene and the derivation disease can be corrected without the risk
with or without insulator elements should of mice from these cells. The animals of insertional mutagenesis. (Blood. 2006;
provide a safe and effective treatment in produce high levels of normal human 108:1183-1188)
humans, some concerns about inser- hemoglobin (HbA) and the pathology as-
tional mutagenesis persist. An ideal cor- sociated with SCD is corrected. Hemato-
rection would involve replacement of the logic values are restored to normal levels
sickle globin gene (␤S) with a normal and organ pathology is ameliorated. © 2006 by The American Society of Hematology

Introduction
Sickle cell disease (SCD) is an autosomal recessive disorder that designed to switch hemoglobins after birth11-13 rather than before
affects a significant proportion (approximately 1 in 500 individu- birth14-19 as observed in animals produced with cosmid, BAC, or
als) of the African-American population. Hispanic, Arabic, Mediter- YAC transgenes. The LCR ␥-␤S transgenic animals are relatively
ranean, and some Asian populations are also affected. More than healthy at birth and then develop severe anemia when the switch to
300 000 individuals worldwide and more than 70 000 in the United HbS is completed at approximately 3 weeks of age. More recently,
States suffer from the disease. The molecular basis for sickle cell we used the same ␥-␤S configuration to produce a knock-in mouse
disease is an A to T transversion in the sixth codon of the human model of sickle cell disease (T.M.R. and T.M.T., unpublished data,
␤-globin gene.1,2 This simple transversion changes a polar glutamic November 2003). These animals complete the switch of human
acid residue to a nonpolar valine in the ␤s-globin chain on the hemoglobins (HbF to HbS) after birth and develop the same severe
surface of HbS (␣2␤S2) tetramers. The valine creates a hydrophobic anemia as the knock-out/transgenic mice during the first week of
projection that fits into a natural hydrophobic pocket formed on Hb life. We derived embryonic stem (ES) cells from these sickle
tetramers after deoxygenation.3,4 The interaction of tetramers knock-in mice for the present studies.
results in the formation of HbS polymers/fibers that cause red blood Two groups have corrected SCD in mouse models by transduc-
cells (RBCs) to become rigid and nondeformable and to occlude tion of hematopoietic stem cells with lentiviral vectors containing
small capillaries.5-10 These vasoocclusive events cause severe antisickling globin genes followed by transplantation of these cells
tissue damage that can result in strokes, splenic infarction, kidney into syngeneic recipients.20,21 Although self-inactivating (SIN)
failure, liver and lung disorders, painful crises, and other complica- lentiviral vectors with or without insulator elements should provide
tions. Cycles of erythrocyte sickling also cause the cells to become a safe and effective treatment for hemoglobinopathies,22 some
fragile, and lysis produces chronic anemia. concerns about insertional mutagenesis persist.23 If viral integra-
Sickle cell disease is normally a relatively benign disorder in the tion inhibits a tumor-suppressor gene or activates an oncogene,
first few months of life because human fetal hemoglobin (HbF) has leukemia can result. Recent studies of children who received a
potent antisickling properties. HbF, which comprises 70% to 90% transplant of retrovirally transduced hematopoietic cells in France
of total hemoglobin at birth, is gradually replaced by HbS during reveal that this outcome can occur.24 An approach that bypasses this
the first few months of life. Rising HbS levels result in the onset of problem is replacement of the sickle globin gene (␤S) with a normal
disease between 3 and 6 months of age. We recently produced a copy of the gene (␤A). In this paper, we demonstrate the replace-
knock-out/transgenic mouse model that mimics this switch from ment of the human ␤S-globin gene with a human ␤A-globin gene by
HbF to HbS. The LCR ␥-␤S transgene in these animals was homologous recombination, and the derivation of mice from these

From the Department of Biochemistry and Molecular Genetics, University of Reprints: Tim M. Townes, Department of Biochemistry and Molecular
Alabama at Birmingham, Schools of Medicine and Dentistry. Genetics, University of Alabama at Birmingham, Schools of Medicine and
Dentistry, Birmingham, AL 35294; e-mail: ttownes@uab.edu.
Submitted February 21, 2006; accepted April 4, 2006. Prepublished online as
Blood First Edition Paper, April 25, 2006; DOI 10.1182/blood-2006-02-004812. The publication costs of this article were defrayed in part by page charge
Supported by grants from the National Heart, Lung, and Blood Institute (T.M.T. payment. Therefore, and solely to indicate this fact, this article is hereby
and T.M.R.). marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734.

L.-C.W., C.-W.S., and T.M.R. contributed equally to this work. © 2006 by The American Society of Hematology

BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4 1183


1184 WU et al BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4

cells. The results demonstrate successful correction of the hemo- phosphate, 0.5 mM EDTA, pH 7.4) and incubated on ice for 15 minutes.
lytic anemia and organ pathology that characterize sickle cell One tenth volume of 10% NaCl was added and the sample was centrifuged
disease in humans. at maximum speed in an Eppendorf microfuge (Hamburg, Germany).
Approximately 1 ␮L of the supernatant was analyzed on an isoelectric
focusing (IEF) gel. IEF was performed using the Isothermal Controlled
Electrophoresis system (Fisher Scientific, Pittsburgh, PA) with precast
Materials and methods agarose IEF gels (RESOLVE from PerkinElmer, Helsinki, Finland).
Hemoglobin bands were quantitated by densitometry with a BioRad
Production of the ⴚ383 ␥-␤A-globin gene construct
(Hercules, CA) GS-800 scanner using Quantity One software (BioRad).
A plasmid containing the human A␥ globin gene with 383 bp of 5⬘ flanking
sequence (4.6 kb total) and a human ␤A globin gene with 815 bp of 5⬘
flanking sequence (4.1 kb total) was digested with SalI to produce an 8.7-kb Hematologic indices and histopathology
⫺383 ␥-␤A DNA fragment. The SalI sites were changed to ClaI by adding Blood was collected from anesthetized animals into Microtainer EDTA
adaptors and the 8.7-kb ClaI fragment was subcloned into pBlueScript. A collection tubes (Becton Dickinson, Franklin Lakes, NJ). RBC count was
herpes simplex virus (HSV) thymidine kinase (TK) gene, which is driven measured on a HemaVet 1700 (CDC Technology, Oxford, CT) hematology
by a phosphoglycerate kinase (PGK) promoter, was digested with XbaI and analyzer. Hemoglobin concentration was determined spectrophotometri-
a 2.0-kb XbaI fragment was isolated. This fragment was subcloned into the
cally after conversion to cyanmethemoglobin with Drabkin reagent (Sigma,
plasmid pTR18 that contains XhoI and SalI sites on either side of XbaI. This
St Louis, MO). Before determining the hemoglobin concentration, red cell
plasmid was digested with XhoI and SalI to generate a 2.0-kb XhoI/SalI
membranes were pelleted at 16 000g for 5 minutes in an Eppendorf
fragment. Another plasmid, containing 1.7 kb of mouse 5⬘ homology, a
microfuge. Packed cell volume (PCV) was measured with a JorVet J503
floxed PGK/Hygromycin (Hygro) gene, and 7 kb of mouse 3⬘ homology,
(Jorgenson Laboratories Systems, Loveland, CO) microhematocrit centri-
was linearized with SalI, and the 13.3-kb DNA fragment was isolated from
fuge. Reticulocyte counts were determined by flow cytometry after staining
an agarose gel. The 2.0-kb XhoI/SalI fragment containing PGK/TK was
with thiazole orange. Urine osmolality was measured with the Wescor
inserted into the SalI site of pTR401. The resulting plasmid was then
Vapro Vapor Pressure Osmometer 5520 (Logan, UT) after food and water
digested with ClaI, which cuts between the mouse 5⬘ homology and the
were withheld from the mice for 16 hours. Tissue preparations from
floxed PGK/Hygro gene, and the 8.7-kb ␥-␤A ClaI fragment was inserted.
This final plasmid containing TK/mouse 5⬘ homology/⫺383 ␥-␤A/floxed the spleen, liver, and kidney were fixed in 70% alcoholic formalin,
PGK/Hygro/mouse 3⬘ homology was digested with multiple enzymes to embedded in paraffin, sectioned, and stained with hematoxylin-eosin stain
verify that the 24-kb plasmid was correct. by standard methods.

ES cell culture and homologous recombination to repair


DNA lesion
Results
The production of a novel, knock-in mouse model of sickle cell disease and
the derivation of ES cells from these mice will be described elsewhere Production of a human ␤A-globin gene targeting construct
(T.M.R. and T.M.T., unpublished data, November 2003). The ⫺383 ␥-␤A
DNA construct was linearized by NotI digestion and electroporated into the We recently produced a knock-in mouse model of sickle cell
knock-in sickle ES cells (h␣/h␣, ⫺1400 ␥-␤S/⫺1400 ␥-␤S). The cells were disease by replacing the mouse ␣-globin genes with a human
plated onto mitomycin C–treated mouse embryonic fibroblasts (MEFs) in ␣-globin gene (h␣/h␣) and by replacing the mouse ␤-globin genes
Dulbecco modification of Eagle medium (DMEM) containing 16.7% fetal with human A␥- and ␤S-globin genes (⫺1400 ␥-␤S/⫺1400 ␥-␤S;
bovine serum (FBS; HyClone, Logan, UT), 1⫻ nucleosides, 2 mM
T.M.R. and T.M.T., unpublished data, November 2003). We also
L-glutamine, 1⫻ nonessential amino acids, 50 IU/mL penicillin, 50 ␮g/mL
streptomycin, 0.1 mM ␤-mercaptoethanol, and 1000 U/mL leukemia
derived ES cells from these knock-in sickle mice. The ES cells are
inhibitory factor (LIF). Positive/negative selection in hygromycin (125 ␮g/mL) homozygous for the human ␣-globin gene (not shown) and
and gancyclovir (2 ␮M) was used to enrich for homologous recombinants.25 homozygous for the A␥- and ␤S-globin genes as indicated in Figure
DNA isolated from individual ES cell colonies was analyzed by polymerase 1A. The A␥-globin gene in the knock-in sickle ES cells contains
chain reaction (PCR). Homologous recombinants were identified with primer 1 1400 bp of 5⬘ flanking sequence; therefore, the genotype is
and primer 2 to identify correct 5⬘ sequences (primer 1 is outside of the vector designated as ⫺1400 ␥-␤S. The targeting construct that we
homology region) and with primers 5 and 6 to identify correct 3⬘ sequences produced for gene replacement contained a human ⫺383 ␥-␤A-
(primer 6 is outside of the vector homology region). PCR with primers 3 and 4
globin gene fragment that is similar to ⫺1400 ␥-␤S except that the
followed by Bsu36I digestion was used to distinguish ␤S and ␤A alleles. Primer
sequences are as follows: primer 1, 5⬘-CTCCTGACTCGGTATCCTGC-3⬘;
␥ gene has only 383 bp of 5⬘ flanking sequence. This-24 kb
primer 2, 5⬘-GAAGTTCTCAGGATCCACATGC-3⬘; primer 3, 5⬘-GATATATCT- replacement vector contained 1.7 kb of mouse 5⬘ flanking se-
TAGAGGAGGGC-3⬘; primer 4, 5⬘-CCAACTTCATCCACGTTCAC-3⬘; primer quence, the ⫺383 ␥-␤A fragment (8.7 kb), a floxed PGK/Hygro
5, 5⬘-CAGAGCTTGGTTGACGGCAATTTCG-3⬘; and primer 6, 5⬘-TGA- gene, 7 kb of mouse 3⬘ flanking sequence, and a PGK/TK gene
GCTCCGGAGGTACCCAGG-3⬘. (Figure 1B).
Positive colonies were electroporated with a cytomegalovirus (CMV)/
Cre plasmid, and individual colonies containing a correctly deleted marker Replacement of the human ␤S-globin gene with the human
gene were identified by PCR. ␤A-globin gene in ES cells derived from knock-in sickle mice

Blastocyst injection and globin protein analysis The targeting construct was electroporated into knock-in sickle ES
cells, and the cells were grown in hygromycin and gancyclovir for
Correctly targeted ES cell lines were injected into C57BL/6 blastocysts. 2 weeks. One hundred thirteen colonies were picked and homolo-
Chimeric males obtained from these injected blastocysts were mated with
gous recombinants were identified by PCR of genomic DNA. In 16
h␣/h␣, ⫺1400 ␥-␤S/m␤ females, and offspring were screened for the
corrected genotypes (⫺383 ␥-␤A/⫺1400 ␥-␤S and ⫺1400 ␥-␤A/⫺1400 (14.2%) of the 113 colonies, the ␤S-globin gene was successfully
␥-␤S; “Results”) by PCR of tail DNA and Bsu36I digestion. replaced with the ␤A-globin gene. In 1 of the 16 colonies,
Fifty microliters of whole blood was washed in 500 ␮L PBS. Cell recombination at the 5⬘ end occurred upstream of the ⫺383 ␥
pellets were resuspended in 100 ␮L lysis solution (5 mM sodium sequence; therefore, the replacement allele contained the
BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4 SICKLE GENE REPLACEMENT 1185

Figure 1. Replacement of the ␤S-globin gene with a ␤A-globin gene in knock-in sickle ES cells. (A) Schematic representation of the human ␤-globin locus, mouse
␤-globin locus, and the human ⫺1400 A␥-␤S knock-in locus in ES cells derived for this study. Arrows indicate DNase I hypersensitive sites (HSs) that mark the locus control
region (LCR). Red and yellow boxes represent functional human and mouse genes, respectively. White boxes represent pseudogenes. Black circles indicate loxP sites. (B)
Schematic representation of gene replacement in knock-in sickle ES cells. The 24-kb replacement vector contains a 2.1-kb PGK/TK marker gene, 1.7 kb of mouse 5⬘ flanking
sequence, a ⫺383 ␥-␤A fragment (8.7 kb), a 1.8-kb floxed PGK/Hygro gene, and 7 kb of mouse 3⬘ flanking sequence. Homologous recombinants were identified by PCR with
primers 1 and 2 to identify correct 5⬘ sequences (primer 1 is outside of the vector homology region) and with primers 5 and 6 to identify correct 3⬘ sequences (primer 6 is outside
of the vector homology region). PCR with primers 3 and 4 followed by Bsu36I digestion was used to distinguish ␤S and ␤A alleles. (C) 5⬘ PCR (primers 1 and 2) and 3⬘ PCR
(primers 5 and 6) from 2 positive homologous recombinant ES cell lines (clones 1 and 2). In clone 1, recombination at the 5⬘ end occurred downstream of the ⫺383 ␥ sequence;
therefore, the replacement allele maintained the ⫺1400 ␥ promoter (2.8-kb PCR product). In clone 2, recombination at the 5⬘ end occurred upstream of the ⫺383 ␥ sequence;
therefore, the replacement allele contained the ⫺383 A␥ promoter (1.8-kb PCR product). Bsu36I digestion of PCR fragments derived with primers 3 and 4 is presented in the
second panel of panel C. ␤A fragments are digested, but ␤S fragments are resistant to digestion.

⫺383 A␥-globin gene promoter (Figure 1B-C, 5⬘ PCR, lane 2; 1-3 and 1-4 are ⫺1400 ␥-␤A/⫺1400 ␥-␤S and animals 2-3 and 2-4
1.8-kb PCR product). In the remaining 15 colonies, recombination are ⫺383 ␥-␤A/⫺1400 ␥-␤S. The 1-1, 1-2, 2-1, and 2-2 animals are
at the 5⬘ end occurred downstream of the ⫺383 ␥ sequence; homozygous sickle mice derived from the same matings. The last 2
therefore, the replacement allele maintained the ⫺1400 ␥ promoter lanes are controls obtained from a human sickle patient (␤S/␤S) and
(Figure 1B-C, 5⬘ PCR, lane 1; 2.8-kb PCR product). Genomic an unaffected individual (␤A/␤A).
DNA from all 16 colonies was analyzed by PCR with primers 3 and Blood samples were obtained from the animals, and hemoly-
4, which amplify both ␤S and ␤A alleles producing 340-bp sates were analyzed on IEF gels. The results are illustrated in
amplicons. These amplicons were digested with Bsu36I, which Figure 2B. The last 3 lanes are control hemolysates from a sickle
does not cut the ␤S allele but digests the ␤A allele into 260- and
80-bp fragments (Figure 1B-C). Homologous recombination at the
3⬘ end of all 16 colonies was correct (Figure 1B-C, 3⬘ PCR; 7.7-kb
PCR product). Cell lines containing the ⫺1400 ␥-␤A replacement
allele or the ⫺383 ␥-␤A replacement allele were transiently
transfected with a CMV/Cre plasmid, and colonies containing a
deletion of the PGK/Hygro marker gene were identified by PCR.
Mice derived from corrected sickle ES cells express high levels
of human HbA

After removal of the marker, ⫺1400 ␥-␤A and ⫺383 ␥-␤A ES cell Figure 2. Genomic DNA and hemoglobin analysis of mice derived from targeted
lines were injected into C57BL/6 blastocysts. Chimeric males ES cell lines 1 and 2. (A) After removal of the PGK/Hygro marker from corrected ES
obtained from these blastocysts were mated with females that were cell clone 1 (⫺1400 ␥-␤A/⫺1400 ␥-␤S) and clone 2 (⫺383 ␥-␤A/⫺1400 ␥-␤S), cells
were injected into C57BL/6 blastocysts. Chimeric males obtained from these
homozygous for human ␣-globin gene knock-in and heterozygous blastocysts were mated with h␣/h␣, ⫺1400 ␥-␤S/m␤ females and offspring were
for human ⫺1400 ␥-␤S knock-in (h␣/h␣, ⫺1400 ␥-␤S/m␤), and screened for the corrected genotypes (⫺1400 ␥-␤A/⫺1400 ␥-␤S and ⫺383 ␥-␤A/
offspring were screened for the corrected genotypes (⫺383 ␥-␤A/ ⫺1400 ␥-␤S) by PCR of tail DNA and Bsu36I digestion. (B) IEF gel of hemolysates
⫺1400 ␥-␤S and ⫺1400 ␥-␤A/⫺1400 ␥-␤S) by PCR of tail DNA from mice identified as sickle and corrected animals in panel A. The last 3 lanes are
human control hemolysates from a sickle patient, an individual with sickle trait, and an
and Bsu36I digestion. Figure 2A demonstrates that animals with unaffected individual. Of interest, the ratio of HbA to HbS in corrected animals mimics
corrected genotypes were obtained from both cell lines. Animals the ratio in humans with sickle trait.
1186 WU et al BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4

Figure 3. Correction of abnormal RBC morphology in


ⴚ383 ␥-␤A–corrected mice. (A) Blood smear of an
h␤A/h␤A control. (B) Blood smear of an h␤S/h␤S sickle
animal with characteristic sickled erythrocytes and a
pronounced reticulocytosis. (C) Blood smear of an h␤A/
h␤S-corrected mouse. No sickled cells were observed in
any field examined. Blood smears were stained with
Wright-Giemsa and the magnification is 100⫻/1.40 NA oil
objective (Nikon Eclipse E800 inverted microscope [Ni-
kon, Tokyo, Japan], Hamamatsu C5810 Color Chilled
3CCD camera [Hamamatsu City, Japan]; Adobe Photo-
shop CS version 8.0 imaging software [San Jose, CA]).
Data from ⫺1400 ␥-␤A–corrected mice are identical to
⫺383 ␥-␤A–corrected animals.

patient, an individual with sickle trait, and an unaffected individual. Amelioration of spleen, liver, and kidney pathology, and
The results in Figure 2B demonstrate that high levels of human restoration of kidney function in mice derived from corrected
HbA are synthesized in red blood cells from 1-3, 1-4, 2-3, and 2-4 ES cells
animals. Of interest, all 4 of these animals mimic the HbA to HbS
␤A/␤S-corrected mice developed little spleen, liver, and kidney
ratio in humans with sickle trait. Individuals with sickle trait
pathology compared with knock-in ␤S/␤S sickle mice. Histologic
present little if any pathology; therefore, these results suggest that
sections of control (␤A/␤A), sickle (␤S/␤S), and corrected (␤A/␤S)
the level of HbA achieved by gene replacement is sufficient to
animals are presented in Figure 4A. The spleens of knock-in sickle
correct the disease.
mice are characterized by a massive expansion of red pulp,
Correction of abnormal RBC morphology and hematologic dramatic pooling of sinusoidal erythrocytes, vasoocclusion, and a
parameters in mice derived from targeted ES cells complete loss of lymphoid follicular structure. In corrected mice,
normal splenic red and white pulp is observed, and virtually no
Blood smears from knock-in sickle (␤S/␤S), corrected (␤A/␤S), and pools of sickle erythrocytes or infarcts are evident. In addition,
control (␤A/␤A) animals are illustrated in Figure 3. Many rigid, splenomegaly is substantially diminished in corrected mice (Figure
elongated cells are observed in the sickle mice (Figure 3B); 4B). The spleens of ␤A/␤S mice weigh approximately 7-fold less
however, no sickled cells are observed in ␤A/␤S-corrected mice or than the spleens of ␤S/␤S animals and are almost the same size as
in ␤A/␤A controls (compare 3A and 3C). The blood smears of spleens of ␤A/␤A control animals.
corrected animals also lack the anisocytosis, poikilocytosis, and The livers of knock-in sickle mice are characterized by focal
polychromasia characteristics of erythrocytes in the sickle mice. areas of necrosis and pronounced congestion of the intrahepatic
The red cells in corrected mice are nearly identical to the cells in vasculature with aggregates of sickled RBCs. Erythroid progeni-
control animals, and this phenotype is similar to human red cells tors are evident in the sinusoids, and this extramedullary hematopoi-
from individuals with sickle trait. esis is indicative of severe anemia. There is also abundant
We also measured hematologic values in the corrected mice and hemosiderin deposition subsequent to Kupffer cell erythrophagocy-
compared these values to the numbers obtained for sickle and tosis. In ␤A/␤S-corrected animals, focal areas of necrosis and
control animals. These data are presented in Table 1. Compared aggregation of sickled erythrocytes are not observed. In addition,
with sickle animals, the corrected mice have marked increases in extramedullary hematopoiesis and hemosiderin deposition are
RBC counts (6.7 ⫾ 1.1 to 11.3 ⫾ 1.0 ⫻ 1012/L [6.7 ⫾ 1.1 to absent. These results demonstrate that organ pathology is signifi-
11.3 ⫾ 1.0 ⫻ 106/␮L]), Hb levels (71 ⫾ 9 to 108 ⫾ 10 g/L cantly ameliorated in mice derived from corrected ES cells.
[7.1 ⫾ 0.9 to 10.8 ⫾ 1.0 g/dL]), and hematocrit levels (.351 ⫾ .033 In the kidneys of knock-in sickle mice, engorgement and
to .417 ⫾ .025 as a proportion of 1 [35.1% ⫾ 3.3% to occlusion of blood vessels with sickled erythrocytes results in
41.7% ⫾ 2.5%]) and a significant reduction in reticulocyte counts vascular, tubular, and glomerular changes. Sequestration and
(.754 ⫾ .039 to .084 ⫾ .026 as a proportion of RBCs occlusion are most obvious at the corticomedullary junctions where
[75.4% ⫾ 3.9% to 8.4% ⫾ 2.6%]). All of the values in the dilated capillaries are easily observed in this region of reduced
corrected mice (␤A/␤S) are similar to values in controls (␤A/␤A). oxygen tension. Reduced medullary blood flow in HbS patients
These data demonstrate that the hematologic defects in sickle cell causes extensive tubular damage that results in hyposthenuria, and
disease can be corrected by replacing one ␤S allele with a ␤A allele this same loss of urine-concentrating ability is observed in the
in ES cells. knock-in ␤S/␤S sickle mice. In contrast, the kidneys of ␤A/␤S-

Table 1. Correction of hematologic pathology and urine concentration defect in mice by homologous recombination
Urine
RBC, Reticulocytes, concentration,
Mice ⴛ 106/␮L Hb, g/dL PCV, % % RDW, % MCV, fL MCH, pg mOsm

h␤A/h␤A control 10.7 ⫾ 0.5 10.5 ⫾ 0.6 41.1 ⫾ 1.9 7.7 ⫾ 2.9 21.7 ⫾ 2.0 38.4 ⫾ 2.0 9.8 ⫾ 0.8 2129 ⫾ 281
h␤S/h␤S sickle 6.7 ⫾ 1.1 7.1 ⫾ 0.9 35.1 ⫾ 3.3 75.4 ⫾ 3.9 32.5 ⫾ 2.1 52.4 ⫾ 3.5 10.6 ⫾ 1.4 768 ⫾ 102
h␤A/h␤S corrected 11.3 ⫾ 1.0* 10.8 ⫾ 1.0* 41.7 ⫾ 2.5* 8.4 ⫾ 2.6* 22.6 ⫾ 1.2* 36.9 ⫾ 2.7* 9.6 ⫾ 1.4 2388 ⫾ 397*

All animals were analyzed at 6 to 8 weeks after birth. Values represent the mean ⫾ SD. Statistical significance was determined for h␤A/h␤S-corrected mice compared with
h␤S/h␤S sickle animals; P values were calculated using a 2-tailed Student t test; n ⱖ 6.
RBC indicates red blood cell; Hb, hemoglobin; PCV, packed cell volume or hematocrit; RDW, red cell distribution width; MCV, mean corpuscular volume; fL, femtoliter; and
MCH, mean corpuscular hemoglobin. To convert RBC counts to SI units, multiply by 106; for Hb, multiply by 10; and for reticulocytes, divide by 100.
*P ⱕ .001.
BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4 SICKLE GENE REPLACEMENT 1187

fragment containing the human A␥-globin gene (5.6 kb) and a


human ␤S-globin gene (4.1 kb). The ␥- and ␤S-globin genes contain
1400 bp and 815 bp of 5⬘ flanking sequence, respectively. Mouse
␣-globin genes were also replaced with a human ␣-globin gene. At
birth, these knock-in sickle mice continue to synthesize significant
amounts of human HbF. During the first week after birth, the
animals switch to more than 99% HbS and develop severe disease.
We linked the human ␥- and ␤S-globin genes in a relatively small
fragment because we previously found that this design resulted in
an HbF to HbS switch that mimicked the switch in human sickle
patients.11-13 Larger fragments derived from cosmids, YACs, and
BACs switch hemoglobins in utero,14-19 and this early switch from
HbF to high levels of HbS results in a high rate of perinatal
mortality. The ␥-␤S fragment in the knock-in sickle mice is
identical to the configuration of the transgene in our previous
knock-out/transgenic sickle mice, and globin gene switching is
similar in these 2 models.
We derived ES cells from these animals and tested a gene
replacement protocol for correcting the disease. This approach
avoids random insertions that are characteristic of viral gene
therapy approaches. Although relatively few insertional mutations
have been observed in viral gene therapy studies, the SCID gene
therapy trials in France demonstrated that leukemic cell clones can
arise from insertional activation of LMO-2,24 and experiments in
nonhuman primates have suggested that insertional inactivation of
BCL-2A1 can result in acute myeloid leukemia.26 The risk of
mutagenesis is a consequence of random insertion of one or more
copies of the viral vector in a large number of cells. If 2 to 3 million
CD34⫹ cells per kilogram of body weight are transduced and
transplanted, a 50-kg patient would receive 100 million cells and,
potentially, 100 million different viral insertions. Although the
number of reported insertional mutations after viral gene therapy
has been low, the large number of insertion sites remains a concern.
The gene replacement strategy described in this paper avoids
the risk of insertional mutagenesis. The defective ␤-globin gene
(␤S) was replaced with a normal copy of the gene (␤A) in ES cells,
and cells derived from a single corrected clone were fully
characterized before transplantation. In the present study, the cells
Figure 4. Amelioration of spleen, liver, and kidney pathology in ⴚ383 ␥-␤A– were transplanted into blastocysts, and hematopoietic stem cells
corrected mice. (A) Spleen, liver, and kidney sections were analyzed at low
(10⫻/0.45 NA objective for spleen and kidney, 40⫻/0.75 NA objective for liver) and
(HSCs) derived from these cells in vivo produced corrected red
high (100⫻/1.40 NA oil objective) magnification. In ⫺383 ␥-␤A–corrected mice, blood cells that did not sickle in recipients; consequently, the
normal splenic red and white pulp is observed, and virtually no pools of sickle anemia and organ pathology of the disease was cured. Of course,
erythrocytes or infarcts are evident. In livers of ⫺383 ␥-␤A animals, focal areas of
necrosis and aggregation of sickled erythrocytes are not observed; also, extramedul-
translation of this approach to human patients would involve in
lary hematopoiesis and hemosiderin deposition are absent. Kidneys of ⫺383 ␥-␤A vitro derivation of HSCs27-29 from corrected, patient-specific ES
mice appear normal and free of the disruptive vascular RBC pooling. All sections cells. This approach would provide corrected, fully characterized,
were stained with hematoxylin-eosin. (B) Correction of splenomegaly in ⫺383
syngeneic HSCs for intravenous transplantation, and red blood
␥-␤A–corrected mice. Data from ⫺1400 ␥-␤A–corrected mice are identical to ⫺383
␥-␤A–corrected animals. cells derived from these HSCs in vivo would not sickle.
The application of this approach to patients with sickle cell
corrected mice appear normal and free of the disruptive vascular disease requires successful derivation of patient-specific ES cells
RBC pooling and hemosiderin deposits observed in the sickle mice. by somatic cell nuclear transfer (SCNT). The recent retractions of 2
Most importantly, urine-concentrating ability is completely re- papers30,31 describing the derivation of human ES cells by this
stored in ␤A/␤S-corrected mice (Table 1). Urine concentrations are technique are a significant setback for the field. However, the
increased from 768 ⫾ 102 mOsm in ␤S/␤S animals to 2388 ⫾ 397 feasibility of the approach has been demonstrated in animal
mOsm in ␤A/␤S mice; urine concentrations in ␤A/␤A controls are models.32,33 Therefore, with additional basic research, there is a
2129 ⫾ 281. These data demonstrate that kidney function is reasonable expectation that patient-specific, nuclear transfer ES
restored in mice derived from corrected ES cells. (ntES) cells can be derived in the future. Correction of the sickle
mutation by gene replacement in ntES cells derived from skin
fibroblasts (or in skin fibroblasts before nuclear transfer) should
Discussion provide a means to produce corrected cells that can be differenti-
ated into HSCs in vitro and transplanted into patients. Red blood
We have recently produced a new mouse model of sickle cell cells derived from these HSCs in vivo will not sickle, and this
disease by replacing the mouse ␤-globin genes with a 9.7-kb DNA treatment should cure the disease.
1188 WU et al BLOOD, 15 AUGUST 2006 䡠 VOLUME 108, NUMBER 4

References
1. Ingram VM. A specific chemical difference be- 14. Paszty C, Brion CM, Manci E, et al. Transgenic two patients after gene therapy for SCID-X1. Sci-
tween the globins of normal human and sickle- knockout mice with exclusively human sickle he- ence. 2003;302:415-419.
cell anaemia haemoglobin. Nature. 1956;178: moglobin and sickle cell disease. Science. 1997; 25. Mansour SL, Thomas KR, Capecchi MR. Disrup-
792-794. 278:876-878. tion of the proto-oncogene int-2 in mouse em-
2. Ingram VM. Gene mutations in human haemoglo- 15. Peterson KR, Clegg CH, Huxley C, et al. Trans- bryo-derived stem cells: a general strategy for
bin: the chemical difference between normal and genic mice containing a 248-kb yeast artificial targeting mutations to non-selectable genes. Na-
sickle cell haemoglobin. Nature. 1957;180:326- chromosome carrying the human beta-globin lo- ture. 1988;336:348-352.
328. cus display proper developmental control of hu- 26. Seggewiss R, Pittaluga S, Adler RL, et al. Acute
3. Wishner BC, Ward KB, Lattman EE, Love WE. man globin genes. Proc Natl Acad Sci U S A. myeloid leukemia associated with retroviral gene
Crystal structure of sickle-cell deoxyhemoglobin 1993;90:7593-7597. transfer to hematopoietic progenitor cells in a rhe-
at 5 A resolution. J Mol Biol. 1975;98:179-194. 16. Strouboulis J, Dillon N, Grosveld F. Developmen- sus macaque. Prepublished on January 26, 2006,
4. Padlan EA, Love WE. Refined crystal structure of tal regulation of a complete 70-kb human beta- as DOI 10.1182/blood-2005-10-4108. (Now avail-
deoxyhemoglobin S: II, molecular interactions in globin locus in transgenic mice. Genes Dev. able as Blood. 2006;107:3865-3867.)
the crystal. J Biol Chem. 1985;260:8280-8291. 1992;6:1857-1864. 27. Zambidis ET, Peault B, Park TS, Bunz F, Civin CI.
17. Greaves DR, Fraser P, Vidal MA, et al. A trans- Hematopoietic differentiation of human embry-
5. Hofrichter J, Ross PD, Eaton WA. Kinetics and
genic mouse model of sickle cell disorder. Nature. onic stem cells progresses through sequential
mechanism of deoxyhemoglobin S gelation: a
1990;343:183-185. hematoendothelial, primitive, and definitive
new approach to understanding sickle cell dis-
ease. Proc Natl Acad Sci U S A. 1974;71:4864- 18. Gaensler KM, Kitamura M, Kan YW. Germ-line stages resembling human yolk sac development.
4868. transmission and developmental regulation of a Blood. 2005;106:860-870.
150-kb yeast artificial chromosome containing the 28. Wang Y, Yates F, Naveiras O, Ernst P, Daley GQ.
6. Crepeau RH, Dykes G, Garrell R, Edelstein SJ.
human beta-globin locus in transgenic mice. Proc Embryonic stem cell-derived hematopoietic stem
Diameter of haemoglobin S fibres in sickled cells.
Natl Acad Sci U S A. 1993;90:11381-11385. cells. Proc Natl Acad Sci U S A. 2005;102:19081-
Nature. 1978;274:616-617.
19. Kaufman RM, Pham CT, Ley TJ. Transgenic anal- 19086.
7. Dykes GW, Crepeau RH, Edelstein SJ. Three-
ysis of a 100-kb human beta-globin cluster-con- 29. Willey S, Ayuso-Sacido A, Zhang H, et al. Accel-
dimensional reconstruction of the 14-filament fi-
taining DNA fragment propagated as a bacterial eration of mesoderm development and expansion
bers of hemoglobin S. J Mol Biol. 1979;130:451-
artificial chromosome. Blood. 1999;94:3178- of hematopoietic progenitors in differentiating ES
472.
3184. cells by the mouse Mix-like homeodomain tran-
8. Noguchi CT, Schechter AN. The intracellular poly- scription factor. Prepublished on January 10,
20. Levasseur DN, Ryan TM, Pawlik KM, Townes
merization of sickle hemoglobin and its relevance 2006, as DOI 10.1182/blood-2005-10-4120. (Now
TM. Correction of a mouse model of sickle cell
to sickle cell disease. Blood. 1981;58:1057-1068. available as Blood. 2006;107:3122-3130.)
disease: lentiviral/antisickling beta-globin gene
9. Brittenham GM, Schechter AN, Noguchi CT. He- transduction of unmobilized, purified hematopoi- 30. Hwang WS, Roh SI, Lee BC, et al. Patient-spe-
moglobin S polymerization: primary determinant etic stem cells. Blood. 2003;102:4312-4319. cific embryonic stem cells derived from human
of the hemolytic and clinical severity of the sick- SCNT blastocysts. Science. 2005;308:1777-
21. Pawliuk R, Westerman KA, Fabry ME, et al. Cor-
ling syndromes. Blood. 1985;65:183-189. 1783.
rection of sickle cell disease in transgenic mouse
10. Eaton WA, Hofrichter J. Hemoglobin S gelation models by gene therapy. Science. 2001;294: 31. Hwang WS, Ryu YJ, Park JH, et al. Evidence of a
and sickle cell disease. Blood. 1987;70:1245- 2368-2371. pluripotent human embryonic stem cell line de-
1266. 22. Puthenveetil G, Scholes J, Carbonell D, et al. rived from a cloned blastocyst. Science. 2004;
11. Ryan TM, Townes TM, Reilly MP, et al. Human Successful correction of the human beta-thalas- 303:1669-1674.
sickle hemoglobin in transgenic mice. Science. semia major phenotype using a lentiviral vector. 32. Rideout WM III, Hochedlinger K, Kyba M, Daley
1990;247:566-568. Blood. 2004;104:3445-3453. GQ, Jaenisch R. Correction of a genetic defect by
12. Behringer RR, Ryan TM, Palmiter RD, Brinster 23. Imren S, Fabry ME, Westerman KA, et al. High- nuclear transplantation and combined cell and
RL, Townes TM. Human gamma- to beta-globin level beta-globin expression and preferred intra- gene therapy. Cell. 2002;109:17-27.
gene switching in transgenic mice. Genes Dev. genic integration after lentiviral transduction of 33. Brambrink T, Hochedlinger K, Bell G, Jaenisch R.
1990;4:380-389. human cord blood stem cells. J Clin Invest. 2004; ES cells derived from cloned and fertilized blasto-
13. Ryan TM, Ciavatta DJ, Townes TM. Knockout- 114:953-962. cysts are transcriptionally and functionally indis-
transgenic mouse model of sickle cell disease. 24. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et tinguishable. Proc Natl Acad Sci U S A. 2006;103:
Science. 1997;278:873-876. al. LMO2-associated clonal T cell proliferation in 933-938.

You might also like