ArticlePDF Available

Effective Retrovirus-Mediated Gene Transfer in Normal and Mutant Human Melanocytes

Authors:

Abstract and Figures

Melanocytes represent the second most important cell type in the skin and are primarily responsible for the pigmentation of skin, hair, and eyes. Their function may be affected in a number of inherited and acquired disorders, characterized by hyperpigmentation or hypopigmentation, consequent aesthetic problems, and increased susceptibility to sun-mediated skin damage and photocarcinogenesis. Nevertheless, the possibility of genetically manipulating human melanocytes has been hampered so far by a number of limitations, including their resistance to retroviral infection. To address the problem of human melanocyte transduction, we generated a melanocyte culture from a patient affected with ocular albinism type 1 (OA1), an X-linked pigmentation disorder, characterized by severe reduction of visual acuity, retinal hypopigmentation, and the presence of macromelanosomes in skin melanocytes and retinal pigment epithelium (RPE). The cultured patient melanocytes displayed a significant impairment in replication ability and showed complete absence of endogenous OA1 protein, thus representing a suitable model for setting up an efficient gene transfer procedure. To correct the genetic defect in these cells, we used a retroviral vector carrying the OA1 cDNA and exploited a melanocyte-keratinocyte coculturing approach. Despite their lower replication rate with respect to wildtype cells, the patient melanocytes were efficiently transduced and readily selected in vitro, and were found to express, process, and properly sort large amounts of recombinant OA1 protein. These results indicate the feasibility of efficiently and stably transducing in vitro not only normal neonatal, but also mutant adult, human melanocytes with nonmitogenic genes.
Content may be subject to copyright.
HUMAN GENE THERAPY 13:947957 (May 20, 2002)
© Mary Ann Liebert, Inc.
Effective Retrovirus-Mediated Gene Transfer in Normal and
Mutant Human Melanocytes
MARIA VITTORIA SCHIAFFINO,
1,2
ELENA DELLAMBRA,
3
KATIA CORTESE,
4
CINZIA BASCHIROTTO,
1
SERGIO BONDANZA,
3
MAURIZIO CLEMENTI,
5
PAOLO NUCCI,
6
ANDREA BALLABIO,
1,7,8
CARLO TACCHETTI,
4
and MICHELE DE LUCA
3
ABSTRACT
Melanocytes represent the second most important cell type in the skin and are primarily responsible for the
pigmentation of skin, hair, and eyes. Their function may be affected in a number of inherited and acquired
disorders, characterized by hyperpigmentation or hypopigmentation, consequent aesthetic problems, and in-
creased susceptibility to sun-mediated skin damage and photocarcinogenesis. Nevertheless, the possibility of
genetically manipulating human melanocytes has been hampered so far by a number of limitations, includ-
ing their resistance to retroviral infection. To address the problem of human melanocyte transduction, we
generated a melanocyte culture from a patient affected with ocular albinism type 1 (OA1), an X-linked pig-
mentation disorder, characterized by severe reduction of visual acuity, retinal hypopigmentation, and the pres-
ence of macromelanosomes in skin melanocytes and retinal pigment epithelium (RPE). The cultured patient
melanocytes displayed a significant impairment in replication ability and showed complete absence of en-
dogenous OA1 protein, thus representing a suitable model for setting up an efficient gene transfer procedure.
To correct the genetic defect in these cells, we used a retroviral vector carrying the OA1 cDNA and exploited
a melanocytekeratinocyte coculturing approach. Despite their lower replication rate with respect to wild-
type cells, the patient melanocytes were efficiently transduced and readily selected in vitro, and were found
to express, process, and properly sort large amounts of recombinant OA1 protein. These results indicate the
feasibility of efficiently and stably transducing in vitro not only normal neonatal, but also mutant adult, hu-
man melanocytes with nonmitogenic genes.
947
OVERVIEW SUMMARY
The skin represents one of the most attractive target tissues
for ex vivo gene therapy, not only for its accessibility, but also
for the availability of advanced skin culture and surgery tech-
niques. Moreover, we previously showed that the main cell
type in the skin, the keratinocyte and its precursor stem cells,
can be efficiently and stably transduced by retroviral vectors
and selected in vitro. However, no effective gene transfer pro-
cedure has been developed yet for the melanocytes, despite
the fact that these cells represent the second physiologically
most important cell type in the skin. We now report the set-
ting up of an improved gene transfer method for normal and
mutant human melanocytes, allowing the expression of non-
mitogenic recombinant proteins in high amounts and in a
uniform and a stable manner. Our findings indicate the fea-
sibility of an ex vivo gene therapy approach for the treatment
of inherited and acquired pigmentation disorders.
1
TIGEM, Telethon Institute of Genetics and Medicine, 20132 Milan, Italy.
2
Present address: DIBIT, Scientific Institute San Raffaele, 20132 Milan, Italy.
3
Laboratory of Tissue Engineering, IDI IRCCS, Istituto Dermopatico dell’Immacolata, 00040 Pomezia (Rome), Italy.
4
Department of Experimental Medicine, Anatomy Section, University of Genoa, 16132 Genoa, Italy.
5
Medical Genetics, Department of Pediatrics, University of Padua, 35128 Padua, Italy.
6
Department of Ophthalmology, San Raffaele Hospital, 20132 Milan, Italy.
7
San Raffaele Faculty of Medicine, 20132 Milan, Italy.
8
Present address: TIGEM, Telethon Institute of Genetics and Medicine, and Second University of Naples, 80131 Naples, Italy.
INTRODUCTION
I
N MAMMALS, pigmentation of the skin, hair, and eyes results
from the presence and distribution of melanins. These black-
brown and yellow-red pigments are exclusively synthesized by
a relatively small subpopulation of highly specialized pigment
cells, including the melanocytes of the skin, eyes, and other tis-
sues, and the retinal pigment epithelium (RPE) (for review see
Quevedo
et al
., 1987; Hearing and Tsukamoto, 1991; King
et
al
., 1995). The melanogenic function of the pigment cells may
be affected in a number of inherited and acquired disorders, re-
sulting in clinical conditions characterized by hyperpigmenta-
tion or hypopigmentation. These can generate serious aesthetic
problems: the cosmetic disfigurement caused, for example, by
vitiligo can generate profound psychological and psychosocial
effects in the affected patients, including depression and social
rejection. Moreover, melanins act as a weak sunscreen, repre-
senting an important defense against ultraviolet radiation.
Therefore, melanocyte malfunctioning can expose the skin to
the damaging effects of sunlight and increase the risk of ma-
lignant melanoma (Sober
et al
., 1991).
The genetic disorders of pigmentation consist of a vast and
heterogeneous collection of pathological conditions. Among
these, albinism represents a group of inherited abnormalities
characterized by primary and specific involvement of the pig-
ment cells of the skin and eyes, which show defective melanin
synthesis and/or distribution. Patients affected with albinism ex-
hibit variable hypopigmentation of the skin and severe devel-
opmental defects of the optic system. The most common forms
of albinism, that is, oculocutaneous albinism (OCA) types 1 and
2, result from alterations of the melanin biosynthetic pathway
with absence or major reduction of melanin (King
et al
., 1995).
In contrast, ocular albinism type 1 (OA1; MIM [Mendelian In-
heritance in Man (McKusick, 1998)] 300500), representing the
most common form of ocular albinism with an estimated preva-
lence of 1:50,000, is thought to arise from abnormal biogene-
sis of melanosomes, that is, the specialized subcellular or-
ganelles of the pigment cells devoted to the synthesis, storage,
and transport of melanins (O’Donnell
et al
., 1976; Garner and
Jay, 1980; Wong
et al
., 1983; Incerti
et al
., 2000).
Ocular albinism is transmitted as an X-linked trait, with car-
rier females showing only minor ocular and skin abnormalities.
Conversely, affected males exhibit the optic changes typical of
all forms of albinism (severe reduction of visual acuity due to
foveal hypoplasia, nystagmus, strabismus, photophobia, iris
translucency, hypopigmentation of the retina, and misrouting
of the optic tracts resulting in loss of stereoscopic vision) and
the presence of giant melanosomes (macromelanosomes) in
skin melanocytes and RPE (O’Donnell
et al
., 1976; King
et al
.,
1995). Progress has been achieved in understanding the mo-
lecular bases for ocular albinism. Indeed, we previously iden-
tified the gene responsible for this disorder and characterized
its protein product as a pigment cell-specific glycoprotein lo-
calized to the melanosomal membrane and displaying features
of G protein-coupled receptors (GPCRs) (Bassi
et al
., 1995;
Schiaffino
et al
., 1996, 1999).
As a first step toward the development of gene therapy ap-
proaches aimed at the correction of pigmentation disorders, we
address here the problem of setting up an effective gene trans-
fer protocol for transducing with nonmitogenic genes not only
normal, but also mutant, melanocytes. For this purpose, we iso-
lated and cultured
in vitro
skin melanocytes obtained from a
patient affected with ocular albinism and showing complete ab-
sence of endogenous OA1 protein. Using OA1 as a physiolog-
ical marker protein, we exploited these cells as a model to de-
velop an extremely efficient gene transfer procedure mediated
by retroviral vectors.
MATERIALS AND METHODS
Microsatellite and sequence analyses of the OA1 gene
Microsatellite markers DXS1223 and DXS7108 were previ-
ously described (Ferrero
et al
., 1995). In addition, we used a
newly identified, highly polymorphic, 19-CA dinucleotide re-
peat (named OA1-CA2) located approximately 1 kb upstream
of exon 1. Oligonucleotide primers flanking the repeat sequence
were OA1-CA2/F (59-TCTTGTGTTGTACTTATGCTGAG)
and OA1-CA2/R (59-GATTACACCACTGCACTCCAG). Be-
cause of the presence of an
Alu
sequence in the region of OA1-
CA2/R, labeling was performed on the F primer only. Poly-
merase chain reaction (PCR) conditions were 30 cycles of 94°C
for 50 sec, 58°C for 50 sec, and 72°C for 40 sec to obtain an
amplification product of 164 bp (with 19 repeats). Exons of the
OA1
gene were amplified and subjected to direct sequencing
analysis as described (Schiaffino
et al
., 1995). Blood and skin
biopsy samples were obtained after all members of the OA1
family had given informed consent.
Melanocyte isolation, culturing, and transduction
Human keratinocytes and melanocytes were isolated from
in
vitro
-reconstituted epidermal sheets and cultivated as described
(De Luca
et al
., 1988; Schiaffino
et al
., 1996). A 2-cm
2
shave
biopsy from the abdominal skin of patient II-3 was used as start-
ing material to isolate the
OA1
-mutant melanocytes. After pu-
rification, melanocytes were cultivated in melanocyte growth
medium: Dulbecco’s modified Eagles medium (DMEM) and
Ham’s F12 medium (2:1 mixture), containing fetal calf serum
(FCS, 5%), insulin (5 mg/ml), adenine (0.18 m
M
), hydrocorti-
sone (0.4 mg/ml), triiodothyronine (2 n
M
), epidermal growth
factor (EGF, 10 ng/ml), basic fibroblast growth factor (bFGF,
1 ng/ml), cholera toxin (CT, 0.1 n
M
), phorbol 12-myristate
13-acetate (PMA, 10 ng/ml), glutamine (4 m
M
), and peni-
cillin–streptomycin (50 IU/ml).
In some experiments, aimed at testing the possibility that
macromelanosome formation could be impaired by growth fac-
tors present in the medium (particularly those known to act
along typical GPCR-mediated pathways) or by absence of the
keratinocyte environment, the patient melanocytes were also
cultured (1) in the absence of CT, which irreversibly activates
Gs protein a chain; (2) in the absence of PMA, which acti-
vates protein kinase C (PKC); (3) in the absence of both CT
and TPA; (4) in the presence of keratinocytes with or without
cholera toxin, or by substituting cholera toxin with the a chain
of melanocyte-stimulating hormone (a-MSH), for up to 2
weeks.
The ages of the donors of human melanocyte strains utilized
in this study were as follows: MK69, 16 years old; MK57, 10
years old; MK106, 40 years old; patient II-3, 5 years old. The
doubling time of the isolated melanocyte cultures was as fol-
lows: normal melanocyte strains MK69, MK57 and MK106,
SCHIAFFINO ET AL.
948
2–4 days; patient melanocytes (either untreated or transduced),
6 days (the transduced patient melanocytes displayed a short-
ening of the doubling time from 6 to 4 days during the initial
passages after infection; however, by the time they were har-
vested for analysis, their replication rate had lowered again to
6 days). Melanosome counts were performed on isolated me-
lanocyte cultures between passages 8 and 17 (MK69, passage
15; MK57, passage 8; MK106 wt, passage 5; MK106 trans-
duced, passage 10; patient untreated, passage 15; patient trans-
duced, passage 17).
The LBSN retroviral vector, carrying the cDNA encoding b-
galactosidase (b-Gal), was previously described (Mathor
et al
.,
1996). The LOA1SN retroviral vector, carrying the OA1 cDNA,
was obtained by cloning the OA1 cDNA into the
Eco
RI and
Xho
I sites of the LXSN retroviral vector (Miller and Rosman,
1989). Preparation of high-titer retroviral supernatants and pro-
ducer cell lines was performed essentially as described (Mathor
et al
., 1996). For retroviral infection, human melanocytes from
normal donors and from the OA1 patient (2 3 10
4
/cm
2
) were
seeded onto a feeder layer composed of lethally irradiated (6000
rads) keratinocytes (8 3 10
4
/cm
2
) and producer Am12/LBSN
or Am12/LOA1SN cell lines (4 3 10
4
/cm
2
) in melanocyte
growth medium, containing PMA (10 ng/ml). After 5 days of
cocultivation, melanocytes were passaged and G418 (7.5
mg/ml) was added for 15 days to select the transduced cells.
Selected cells were used for further molecular analysis.
In the melanocyte–keratinocyte coculture assays, transduced
melanocytes (5 3 10
2
/cm
2
) and normal human keratinocytes
(5 3 10
3
/cm
2
) were cocultivated onto a feeder layer of lethally
irradiated 3T3-J2 cells in keratinocyte growth medium: DMEM
and Ham’s F12 medium (2:1 mixture), containing fetal calf
serum (FCS, 10%), insulin (5 mg/ml), adenine (0.18 m
M
), hy-
drocortisone (0.4 mg/ml), triiodothyronine (2 n
M
), epidermal
growth factor (EGF, 10 ng/ml), CT (0.1 n
M
), glutamine (4 m
M
)
and penicillinstreptomycin (50 IU/ml). To determine the lo-
cation and the distribution of melanocytes within the reconsti-
tuted epidermal sheet, confluent cultured epithelia were de-
tached from the culture vessel with the neutral protease Dispase
II, stained with b -galactosidase or dihydroxyphenylalanine
(DOPA), and subjected to histological analysis (De Luca
et al
.,
1988).
Northern, Western, and immunofluorescence analyses
Northern blot analysis was performed as described, using the
OA1 cDNA as a probe (Bassi
et al
., 1995). Western and im-
munofluorescence analyses were performed essentially as de-
scribed (Schiaffino
et al
., 1996, 1999; d’Addio
et al
., 2000).
Protein extracts from normal, patient, and transduced
melanocytes were separated on a sodium dodecyl sulfate
(SDS)–7.5% polyacrylamide gel and transferred to a polyvinyli-
dene difluoride (PVDF) membrane sheet (Hybond-P; Amer-
sham Pharmacia Biotech, Piscataway, NJ), using the Mini-
PROTEAN and the Mini Trans-Blot apparatus (Bio-Rad,
Hercules, CA). Visualization of antibody binding was carried
out with Enhanced ChemiLuminescence Plus (Amersham, Ar-
lington Heights, IL) according to the manufacturer’s instruc-
tions. For immunofluorescence colocalization studies,
melanocytes were cultured on plastic coverslips and fixed in
methanol at 220°C. Affinity-purified anti-OA1 antibody W7
was previously described (Schiaffino
et al
., 1996) and used at
0.5 and at 1.5 mg/ml for Western and immunofluorescence
analyses, respectively. The monoclonal antibody (MAb)
HMB45 against Pmel17 (Dako, Glostrup, Denmark) was used
at a 1:80 dilution for immunofluorescence. Fluorescein isoth-
iocyanate (FITC)-conjugated goat anti-mouse and tetramethyl
rhodamine isothiocyanate (TRITC)-conjugated pig anti-rabbit
immunoglobulins (Dako) were used as secondary antibodies.
Electron microscopy analysis and morphometry
For ultrastructural analysis, normal and OA1 patient
melanocytes were fixed with 2.5% glutaraldehyde in cacody-
late buffer, postfixed in osmium tetroxide, dehydrated through
a graded ethanol series, and embedded in LX112 (Polysciences,
Warrington, PA). Ultrathin sections were stained with uranyl
acetate and lead citrate, and analyzed with EM10C or EM902A
electron microscopes (Zeiss, Thornwood, NY). Melanosome
counts were performed on an average of 10 and 20 cells for
normal and OA1 patient melanocytes, respectively. Counting
included all visible melanosomes, excluding those organized as
aggregates surrounded by a single membrane.
RESULTS
Affected individuals of the OA1 family carry a
frameshift within the OA1 gene
The pedigree of the OA1 family analyzed in this study is
shown in Fig. 1. Complete ocular examination of all family
members was performed. Individual II-1 was examined at the
age of 24 years. Visual acuity for distance, with correction, was
20/100 (20/60 for near vision). The patient showed nystagmus
and iris translucency at slit lamp examination. Fundus obser-
vation disclosed prominent choroidal vessels, indicating hy-
popigmentation of the RPE, and absence of the foveal reflex.
Histological and ultrastructural examinations of a skin bioptic
sample from the patient revealed the presence of typical
macromelanosomes. Individual II-3 was examined at the age of
5 years. Visual acuity for distance, with correction, was 20/200
(20/80 for near vision). The patient also showed nystagmus, iris
translucency, and fundus hypopigmentation, with marked at-
tenuation of the foveal reflex.
Individual I-2 displayed visual acuity of 20/20 and a pattern
of fundus depigmentation consistent with a carrier status for oc-
ular albinism. Ocular examinations of individuals I-1 and II-2
were unremarkable. To confirm the diagnosis of ocular albinism
type 1 at the molecular level, we performed both linkage and
mutation analyses, using DNA samples obtained from fresh
blood of all family members. Linkage analysis was performed
with microsatellite markers located adjacent or internal to the
OA1
gene and showed cosegregation of the disease with the
OA1
locus (Fig. 1). Finally, the direct sequencing of PCR-am-
plified
OA1
exons revealed the presence of a four-nucleotide
deletion at the end of exon 1 in the affected members of the
family, that is, II-1 and II-3 (252del4; Fig. 2A, PT).
OA1 protein is undetectable in OA1
patient melanocytes
The
OA1
gene is expressed, at high levels, only in
melanocytes and RPE, consistent with the clinical phenotype
GENE TRANSFER IN HUMAN MELANOCYTES
949
of the disease. Therefore, to determine the effect of the 252del4
mutation on the OA1 mRNA and protein we obtained a skin
biopsy from patient II-3 and generated a melanocyte culture.
Although the patient melanocytes displayed a lower replication
rate with respect to wild-type cells (the mean population dou-
bling time was 6 days instead of 2–4 days, respectively), they
were isolated and successfully passaged more than 20 times.
The patient melanocytes were initially subjected to Northern
blot analysis with the OA1 cDNA, revealing that the size and
abundance of the OA1 mRNA are normal in these cells (Fig.
2B, PT).
As illustrated in Fig. 3A, the 252del4 mutation results in a
frameshift that could theoretically give rise to two truncated
protein products: a short peptide corresponding to the N termi-
nus and the first transmembrane domain of OA1 up to residue
65, plus 19 unrelated amino acids; and an N terminus-truncated
form of OA1, lacking the first 84 residues and starting from the
second in-frame ATG.
In vitro
studies, performed with the anti-
OA1 antibody (directed against the C terminus of OA1) and a
recombinant construct missing the first ATG, showed that the
N terminus-truncated mutant could actually be produced in
transfected HeLa cells. However, the truncated protein was not
correctly sorted to the lysosomes, displaying a reticular pattern
consistent with retention in the endoplasmic reticulum (ER)
(data not shown).
To test for the presence of a truncated form of OA1 in the
patient melanocytes, we performed Western and immunofluo-
rescence analyses with the anti-OA1 antibody (Fig. 3B, WT and
PT, and not shown). We did not obtain any specific signal by
either method, suggesting that, if alternative start codons down-
stream of the mutation are being used, they produce unstable
proteins not detectable by Western blot and immunofluores-
cence. This interpretation is in agreement with our data indi-
cating that, when expressed at physiological levels, mutant OA1
proteins retained in the ER are rapidly degraded (d’Addio
et
al
., 2000). Conversely, because antibodies against the N termi-
nus of OA1 are not available, we were unable to evaluate the
expression and stability of the theoretical C terminus-truncated
mutant of OA1.
OA1 patient melanocytes display an excess
of mature melanosomes
By light microscopy analysis, the patient melanocytes dis-
played a normal bipolar morphology and an overall appearance
similar to wild-type cells. To look for the presence of subcel-
lular abnormalities and particularly of macromelanosomes, we
performed extensive ultrastructural examination studies. Sur-
prisingly, the cultured patient melanocytes did not show any
structural abnormalities of melanosomes with respect to wild-
type cells, despite the huge number of samples analyzed and
despite the several culture conditions utilized (see Materials and
Methods). Indeed, both normal and patient melanocytes showed
the presence of normal melanosomes at all stages of matura-
tion, and occasionally of melanosomal aggregates. This incon-
sistency between
in vivo
versus cultivated melanocytes cannot
be attributed to patient-based variability. Indeed, we also ana-
lyzed a melanocyte culture obtained from the affected brother
of patient II-3, that is, patient II-1 (the poor viability of this lat-
ter culture did not allow us to use it for further investigation
and transduction). We found that, although examination of a
skin biopsy from patient II-1 revealed the presence of typical
macromelanosomes (see the description of the OA1 family), no
structural abnormalities of melanosomes were observed in the
cultured melanocytes obtained from this same patient.
Nevertheless, a major difference between normal and patient
melanocytes emerged by considering the number of melanosomes
and their maturation stage. Comparing the ratios between mature
SCHIAFFINO ET AL.
950
FIG. 1. Pedigree and microsatellite analysis of the OA1 fam-
ily. DXS 1223 and DXS 7108 are microsatellite markers flank-
ing the
OA1
locus on the telomeric side and on the centromeric
side, respectively. OA1-CA2 represents a novel marker located
within the
OA1
gene (see Materials and Methods for details).
The analysis shows cosegregation of the disease with the
OA1
locus.
FIG. 2. Genomic sequence of the
OA1
gene (A) and North-
ern blot analysis of the
OA1
transcript (B) in a normal indi-
vidual (WT) and in patient II-3 (PT). (A) The patient carries a
deletion of four bases within the coding region of exon 1 of the
OA1
gene (252del4). (B) Northern analysis, performed using 4
mg of total RNA from wild-type melanocytes (WT) and 2 mg
of total RNA from patient melanocytes (PT), reveals that the
OA1 mRNA expressed by patient melanocytes is similar in size
and abundance to that expressed by wild-type cells.
(stage IV, fully pigmented) versus immature (stage II–III, non-
pigmented or partially pigmented) melanosomes, we found a
higher prevalence of mature melanosomes in the patient
melanocytes with respect to wild-type cells. Table 1 shows the
melanosome counts performed in three independent normal me-
lanocyte strains (MK69, MK57, and MK106) and in the patient
melanocytes. Normal melanocytes, obtained from white subjects
with type III–IV skin complexions (as the patient), displayed a
mature/immature melanosome ratio ranging between 0.1 and 0.8,
with an average of 0.46 (Table 1, bottom row, average of wild
type). No major variations were observed in cultures of different
passage number or obtained from donors of different ages (see
Materials and Methods). In contrast, in the patient melanocytes
the mature melanosomes represented the vast majority of the or-
ganelles, with a mean mature/immature melanosome ratio of 6.99
(Table 1, bottom row, patient untreated).
Normal and mutant melanocytes can be
efficiently and stably transduced
Transduction of normal human melanocytes
. Many pigmen-
tation disorders, including albinism and xeroderma pigmento-
sum, result from the loss-of-function of the involved genes and
might therefore be considered as candidates for classic re-
placement gene therapy approaches. However, a major prereq-
uisite to the genetic correction of pigmentation disorders is rep-
resented by the availability of effective gene transfer procedures
for normal and mutant human pigment cells. To set up an effi-
cient transduction protocol for human melanocytes, we initially
utilized wild-type cells and infected them with a Moloney
murine leukemia virus-derived retroviral vector carrying the
bacterial b-galactosidase cDNA (LBSN) (Mathor
et al
., 1996).
In the search for an effective transduction strategy, we rea-
soned that when grown in close contact with keratinocytes, hu-
man melanocytes proliferate at substantially higher rates (the
mean population doubling time becomes 24 hr with respect to
the 2–4 days required by isolated melanocytes), undergo proper
morphological and functional (melanin synthesis) differentia-
tion, and physiologically migrate within the basal layer of the
cultured epidermis (De Luca
et al
., 1988). The ability of ker-
atinocytes to promote melanocyte growth and differentiation
could facilitate melanocyte transduction as well. Therefore, we
cocultured normal human melanocytes with lethally irradiated
keratinocytes and infected them with the retroviral vector
LBSN. The highest transduction efficiency (4050%) was ob-
tained when melanocytes were seeded for at least 5 days on a
feeder layer composed of lethally irradiated keratinocytes and
producer GP1envAm12 cells (2:1 mixture; see Materials and
Methods for details).
After infection, melanocytes were passaged and geneticin
was added for 15 days to select the transduced cells. By this
treatment, the transduction efficiency reached 95–100% (Fig.
4A) and this value was maintained during the following culti-
vation without geneticin, demonstrating that normal neonatal
human melanocytes can be efficiently and stably transduced
in
vitro
with a nonmitogenic marker gene. Finally, to evaluate
whether the transduced melanocytes maintained the proper or-
ganization in the reconstituted epithelial sheet, we cocultured
human keratinocytes with LBSN-transduced melanocytes. We
found that the transduced melanocytes were associated with ke-
ratinocyte colonies like wild-type cells (Fig. 4B) and, after
colony fusion and epidermal sheet reconstitution, were physi-
ologically organized in the basal layer and maintained a nor-
mal melanocyte/keratinocyte ratio (Fig. 4C).
Transduction of OA1 patient melanocytes
. To transduce the
patient melanocytes, we took advantage of the same infection
protocol as described above, with the exception that in this case
GENE TRANSFER IN HUMAN MELANOCYTES
951
FIG. 3. Theoretical and biochemical consequences of the
252del4 mutation on the OA1 protein. (A) Schematic repre-
sentation of the truncated OA1 proteins that could theoretically
be translated by the patient mRNA starting from the first (phys-
iological) and second in-frame ATGs. The predicted heptahe-
lical topology of the wild-type OA1 protein is used as frame-
work. Vertical rectangles (I to VII), transmembrane a helices;
e
1–3
and i
1–3
, hydrophilic lumenal and intracellular loops; N and
C, N terminus and C terminus, respectively; solid circles,
residues encoded by the mutant mRNA following the
frameshift. (B) Western blot analysis of normal melanocytes
(WT), and of patient melanocytes before (PT) and after (PT-
LOA1SN) infection with the LOA1SN retroviral vector. Ar-
rows, 60-kDa glycosylated form and 45- to 48-kDa unglyco-
sylated doublet of the OA1 protein; asterisk, unrelated protein
cross-reacting with the anti-OA1 antibody. A comparable
amount of OA1 protein is detected in 25 mg and in 0.6 mg of
extract from wild-type melanocytes and from LOA1SN-trans-
duced patient melanocytes, respectively, indicating that the lat-
ter express approximately 40 times more OA1 protein than the
former.
SCHIAFFINO ET AL.
952
TABLE 1. MELANOSOM E COUNTS IN NORMAL, PATIENT, AND TRANSDUCED MELANOCYTES
Melanocyte culture
a
Average
MK69 MK57 MK106 MK106 MK106 of wild Patient Patient
(wt) (wt) (wt) LXSN LOA1SN type untreated LOA1SN
Melanosomes per cell
b
125 204 182 152 178 171 354 309
Mature melanosomes
c
012 068 081 064 065 054 304 191
Immature melanosomes
d
113 136 101 088 113 117 050 118
Mature versus immature 0.1 6 0.09 0.57 6 0.25 0.8 6 0.48 0.73 6 0.24 0.63 6 0.38 0.46 6 0.42 6.99 6 3.67 1.64 6 0.60
melanosomes
e
a
MK69, MK57, and MK106, three independent normal human melanocyte strains; MK106-LXSN, normal melanocytes trans-
duced with the empty retroviral vector LXSN; MK106 LOA1SN and patient LOA1SN, normal and patient melanocytes trans-
duced with the retroviral vector LOA1SN, respectively.
b
The number of melanosomes per cell was calculated as the arithmetic average of the counts obtained in 1020 independent
sections from different cells of the same melanocyte culture.
c
Mature melanosomes: stage IV, fully pigmented melanosomes (mean number per cell).
d
Immature melanosomes: stage II–III, nonpigmented or partially pigmented melanosomes (mean number per cell).
e
The mature/immature melanosome ratio, represented in the figure as a histogram, was calculated as the arithmetic average of
the ratios obtained in each independent section analyzed, with the standard deviation.
FIG. 4. Transduction of normal human melanocytes with the LBSN retroviral vector. (A) b-Gal staining of the melanocyte cul-
ture after transduction and G418 selection. All visible cells appear to express the b-Gal enzyme. (B) LBSN-transduced melanocytes
associated with a keratinocyte colony. (C) After colony fusion and epidermal sheet reconstitution, the LBSN-transduced
melanocytes maintain a normal melanocyte/keratinocyte ratio, as previously observed with wild-type melanocytes. Original mag-
nification: 3400.
FIG. 5. Immunofluorescence analysis of patient melanocytes transduced with the LOA1SN retroviral vector. The recombinant
OA1 protein is detected by anti-OA1 antibody (OA1), whereas the melanosomal marker Pmel-17/gp100 is visualized by MAb
HMB45 (gp100). (A) Comparison between staining for OA1 and Pmel-17/gp100 in a representative field at lower original mag-
nification (3400), showing that virtually all G418-selected melanocytes are expressing the recombinant OA1 protein. Note that
in contrast to OA1, which is found only at an intracellular level, Pmel-17/gp100 is also detected at the plasma membrane, as pre-
viously reported. P.O., Phase optics. (B) Two examples of colocalization between OA1 and Pmel-17/gp100 at higher magnifi-
cation (31000), indicating that the recombinant OA1 protein is sorted to the melanosomes as the endogenous protein in wild-
type cells. The strong perinuclear staining obtained with the anti-OA1 antibody is probably due to accumulation of highly
overexpressed protein in the Golgi region (the recombinant OA1 in the transduced melanocytes is expressed about 40 times more
that the endogenous protein in normal melanocytes; see Fig. 3B). m., Merge.
FIG. 4
FIG. 5
the retroviral vector LOA1SN, carrying the OA1 cDNA, was
used. Because endogenous OA1 protein was undetectable in
the patient melanocytes, we could use the anti-OA1 antibody
to verify the reconstitution of OA1 expression, physiological
processing, and subcellular distribution in the transduced
cells. As shown in Fig. 3B, Western blot analysis of extracts
from the patient melanocytes after transduction with the
LOA1SN retroviral vector revealed the reappearance of both
the 60-kDa fully glycosylated form and the 45- to 48-kDa
unglycosylated polypeptides of the OA1 protein [Fig. 3B,
PT(LOA1SN)]. Serial dilutions of the LOA1SN-transduced
melanocyte extracts showed that these cells express OA1 at
a much higher level than wild-type cells, possibly because of
the strength of the long terminal repeat (LTR) promoter com-
pared with the OA1 promoter, and/or to multiple proviral in-
tegrations (Fig. 3B).
Moreover, immunofluorescence analysis using bright-field
and melanocyte markers for comparison revealed that trans-
duction efficiency was about 80–90% before the selection and
reached 100% after the addition of geneticin (not shown and
Fig. 5A). The recombinant OA1 protein was found to be sta-
bly expressed at high levels for several passages after infection
(at least six passages) and to partially colocalize with the
melanosomal marker gp100/Pmel-17 (Fig. 5B), thus displaying
a distribution pattern similar to that of the endogenous protein
in wild-type melanocytes. Finally, transduction of the patient
melanocytes with the LOA1SN retroviral vector was sufficient
to substantially revert the melanosomal maturation phenotype,
characterized by predominance of mature melanosomes, re-
ducing the mature/immature melanosome ratio toward normal
values (Table 1, from 6.99 in patient untreated to 1.64 in pa-
tient LOA1SN). Instead, no significant modification of the ma-
ture/immature melanosome ratio, or of melanosome morphol-
ogy, was observed in normal melanocytes transduced with the
empty LXSN vector or with the LOA1SN vector (Table 1,
MK106 LXSN and MK106 LOA1SN, respectively).
The LOA1SN retroviral vector appeared to confer a moder-
ate growth advantage to the patient melanocytes. During the
initial passages after infection, the LOA1SN-transduced patient
melanocytes displayed a shortening of doubling time (from 6
to 4 days in untreated and transduced cells, respectively), which
possibly facilitated cell recovery and selection. However, such
advantage was apparently lost after a few passages, so that, by
the time of harvesting for analysis, the replication rate of the
transduced melanocytes had lowered again to 6 days. Instead,
we could not obtain successful transduction of the patient
melanocytes with the empty LXSN retroviral vector, probably
because of the impaired replication ability of these cells. Nev-
ertheless, the inability of both the LOA1SN and LXSN vectors
to influence melanosome maturation in normal melanocytes
supports the specificity of the phenotype rescue obtained with
the LOA1SN vector in the patient melanocytes.
DISCUSSION
Transduction of normal and mutant
human melanocytes
The skin certainly represents one of the most attractive tar-
get tissues for gene therapy. Over the last 50 years, major ad-
vances have been achieved in the development of skin culture
and surgery techniques aimed at the repair of massive full-thick-
ness burns. Large sheets of stratified squamous epithelium can
be obtained in a limited amount of time from small bioptic sam-
ples and are routinely utilized for autologous grafting in pa-
tients suffering from large skin and mucosal defects (Gallico
et
al
., 1984; Pellegrini
et al
., 1997, 1999; Rama
et al
., 2001).
Moreover, we have successfully utilized cultured epidermal au-
tograft, bearing a controlled and physiological melanocyte/ke-
ratinocyte ratio, for the treatment of “stable” vitiligo (Guerra
et
al
., 2000). Finally, we have previously shown that the main cell
type in the skin, the keratinocyte and its precursor stem cells,
can be efficiently and stably transduced by retroviral vectors
and selected
in vitro
(Mathor
et al
., 1996; Dellambra
et al
.,
1998). These findings demonstrate that, at least in principle,
ex
vivo
gene therapy approaches using the skin as the target tis-
sue may be feasible for the correction of cutaneous as well as
noncutaneous diseases.
The melanocytes represent the second physiologically most
important cell type in the skin (with a mean rate of 1:35 with
respect to keratinocytes). They have protective and aesthetic
functions, but can also generate a common and extremely se-
vere type of cancer, malignant melanoma. Therefore, the trans-
duction of melanocytes represents a key step for the develop-
ment of therapeutic approaches aimed at the correction of skin
pigmentation disorders, involving melanocytes alone, such as
oculocutaneous albinism, or both melanocytes and ker-
atinocytes, such as xeroderma pigmentosum (XP). Indeed, XP
is an extremely severe genetic disorder characterized by muta-
tion of the genes involved in excision repair of damaged DNA.
Patients with this disorder show hypersensitivity to ultraviolet
rays, with an incidence of squamous and basal cell carcinomas
and melanomas over 2000 times greater than in the normal pop-
ulation. Therefore, both keratinocyte and melanocyte gene cor-
rection would be required for the treatment of this disorder (Car-
reau
et al
., 1995; Zeng
et al
., 1997).
Gene transfer or transgenesis approaches have been widely
utilized in mice and other rodents to perform biological stud-
ies on melanocyte function and development (Kucera
et al
.,
1996; Dunn
et al
., 2000), and to correct various types of ge-
netic or acquired pigmentation disorders, including the melan-
ocyte-derived cancer malignant melanoma (Hirschowitz
et al
.,
1998). Rescue of the hypopigmented phenotype in different
kinds of albinism has been accomplished by several approaches.
Melanocyte cultures, generated from pink-eyed dilution and
brown mice, have been transduced with the corresponding
genes by using LipofectAMINE reagents and retroviruses, re-
spectively (Bennett
et al
., 1990; Sviderskaya
et al
., 1997). Mu-
tations of the tyrosinase gene have been complemented in al-
bino mouse melanocytes and skin by RNA–DNA
oligonucleotide strategies (Igoucheva and Yoon, 2000), in al-
bino mice by cell-type-directed gene targeting
in utero
(Dunn
et al
., 2001), and in albino rabbits by yeast artificial chromo-
some transgenesis (Brem
et al
., 1996).
However, the possibility of using genetically modified hu-
man melanocytes to correct human pigmentation disorders has
been hampered so far by a number of limitations, including the
resistance of these cells to retroviral infection compared with
keratinocytes, possibly depending on their lower replication
abilities. Indeed, it was previously shown that normal human
melanocytes can be transduced with retroviral vectors, although
SCHIAFFINO ET AL.
954
with low efficiency (Coleman and Lugo, 1998; Hamoen
et al
.,
2001). Coleman and Lugo (1998) transduced human
melanocytes with a bFGF-retrovirus or an empty retrovirus, re-
porting yields of 5–10 or 20–60 G418-resistant colonies per in-
fection, respectively. Hamoen
et al
. (2001) transduced human
melanocytes with a hepatocyte growth factor (HGF)-retrovirus,
achieving an efficiency of 6%. Higher transduction efficiencies
were obtained with adenoviral vectors; however, the episomal
nature of adenoviral replication makes such vectors unsuitable
for the gene therapy of a highly self-renewing tissue like the
skin (Nesbit
et al
., 1999). Thus, the possibility of efficiently
and stably transducing human pigment cells, particularly if car-
rying mutations that may affect their viability and growth ca-
pacity and using nonmitogenic genes, remains to be established.
We obtained a melanocyte culture from a 5-year-old indi-
vidual affected with ocular albinism type 1. The patient
melanocytes did not express any detectable OA1 protein and,
as often observed with mutant cells, displayed a significantly
impaired replication ability compared with wild-type cells. Thus
they represented a suitable model for setting up an efficient
gene transfer procedure. For this purpose, we took advantage
of a melanocyte–keratinocyte coculturing approach and trans-
duced the patient melanocytes with the retroviral vector
LOA1SN, carrying the OA1 cDNA. Consequently, we could
evaluate both the efficiency of the transduction protocol and re-
constitution of the physiological processing, targeting, and ac-
tivity of the melanocyte-specific protein OA1 in human pig-
ment cells.
Our results showed that the LOA1SN-transduced patient
melanocytes were able to express large amounts of recombi-
nant OA1 protein and to sustain its correct processing and tar-
geting to the melanosomes. Moreover, the transduced cells dis-
played a substantial recovery from their aberrant predominance
of mature melanosomes, regaining a normal mature/immature
melanosome ratio. The recombinant OA1 protein was found to
be expressed at high levels in 80–90% of the infected
melanocytes before selection and in virtually 100% of the cells
after selection with G418. OA1 expression was maintained for
several passages after infection, indicating that the transgene is
stably integrated in the melanocyte genome and remains func-
tional during the subsequent doublings of the cells. Although
we obtained only a moderate and transient increase in the repli-
cation rate of LOA1SN-transduced patient melanocytes, the
presence of a more powerful and effective growth advantage in
other systems would probably increase the recovery and facil-
itate the selection of the genetically corrected melanocytes.
In summary, we have developed an efficient gene transfer
procedure for human melanocytes, allowing the expression of
nonmitogenic recombinant proteins in high amounts, and in a
uniform and stable manner. We showed that pure cultures of
normal or mutant melanocytes can be transduced, selected, and
eventually used together with keratinocytes for the generation
of genetically modified epidermal sheets suitable for grafting
onto patients. Our results demonstrate the feasibility of an
ex
vivo
gene therapy approach for the correction of inherited and
acquired disorders involving pigment cells of the skin.
The melanosomal phenotype in ocular albinism type 1
The culture conditions used for growing melanocytes
in vitro
are different with respect to those used for intact skin. As a con-
sequence, normal human melanocytes in culture can exhibit var-
ious types of melanosomal abnormalities (Breathnach
et al
.,
1988). Nevertheless, a number of melanocyte strains and lines
have been obtained previously from mice (and humans) affected
with various forms of albinism, with preservation of the original
phenotypes (Park
et al
., 1993; Zhao
et al
., 1994; Bennett and
Sviderskaya, 1996). Although somewhat unexpected, the lack of
typical macromelanosomes in the OA1 patient melanocytes ap-
pears consistent with the variable expressivity (within the same
cell and in different cell types and patients) and with the non-
specificity displayed
in vivo
by this particular melanosomal phe-
notype (O’Donnell
et al
., 1976; Garner and Jay, 1980; Wong
et
al
., 1983; King
et al
., 1995; Schnur
et al
., 1998). Thus, the de-
ficiency of OA1 appears neither sufficient nor necessary by it-
self to determine the manifestation of the macromelanosomal
phenotype, which instead might depend on multiple factors giv-
ing rise to (or preventing) this abnormality in different patho-
logical conditions. On the other hand, the role of macrome-
lanosomes in the pathogenesis of ocular albinism remains to be
established (Incerti
et al
., 2000).
Despite the absence of macromelanosomes, we noticed a sur-
prisingly high prevalence of mature melanosomes in the OA1
patient melanocytes with respect to wild-type cells and a sig-
nificant rescue of this abnormality after transduction with the
LOA1SN retroviral vector (Table 1). These results suggest that
the dysregulation of melanosome biogenesis caused by the de-
ficiency of OA1 might manifest with alternative phenotypes,
presence of macromelanosomes or prevalence of mature
melanosomes,
in vivo
and
in vitro
, respectively. Consistent with
our previous studies on the
Oa1
knockout (Incerti
et al
., 2000),
these findings further support the idea that OA1 could act as a
negative regulator of melanosome maturation, by preventing
melanosome overgrowth and/or uncontrolled melanin deposi-
tion.
ACKNOWLEDGMENTS
We thank Dr. V. Marigo for critical reading of the manu-
script. This work was supported by generous donations from
Vision of Children Foundation-San Diego (to M.V.S. and A.B.),
Telethon-Italy (Telethon grants F.3 to M.V.S., A.106 and B.53
to M.D.L., and E0942 to C.T.), CNR (target project “Biotech-
nology to C.T.), and MURST (to C.T.). We also warmly thank
all members of the OA1 family involved in this study, as with-
out their collaboration this work would not have been possible.
REFERENCES
BASSI, M.T., SCHIAFFINO, M.V., RENIERI, A., DE NIGRIS, F.,
GALLI, L., BRUTTINI, M., GEBBIA, M., BERGEN, A.A.B.,
LEWIS, R.A., and BALLABIO, A. (1995). Cloning of the gene for
ocular albinism type 1 from the distal short arm of the X chromo-
some. Nat. Genet. 10, 13–19.
BENNETT, D.C., HUSZAR, D., LAIPIS, P.J., JAENISCH, R., and
JACKSON, I.J. (1990). Phenotypic rescue of mutant brown
melanocytes by a retrovirus carrying a wild-type tyrosinase-related
protein gene. Development 110, 471–475.
BENNETT, D.C., and SVIDERSKAYA, E.V. (1996). Advances in im-
mortalization of cultured melanocytes and melanoblasts. In The Pig-
mentary System and Its Disorders. J.J. Nordlund, V.J. Hearing, R.A.
GENE TRANSFER IN HUMAN MELANOCYTES
955
King, and J.P. Ortonne, eds. (Oxford University Press, New York)
pp. 165–174.
BREATHNACH, A.S., PÄTZOLD, H.C., ROBINS, E.J., BHASIN,
Y.D., ETHRIDGE, L., and NAZZARO-PORRO, M. (1988). Trans-
mission and scanning electron microscopy and freeze–fracture repli-
cation of normal human melanocytes and human melanoma cells in
tissue culture. Pigment Cell Res. 1, 315–325.
BREM, G., BESENFELDER, U., AIGNER, B., MULLER, M., LIEBL,
I., SCHUTZ, G., and MONTOLIU, L. (1996). YAC transgenesis in
farm animals: Rescue of albinism in rabbits. Mol. Reprod. Dev. 44,
5662.
CARREAU, M., QUILLIET, X., EVENO, E., SALVETTI, A.,
DANOS, O., HEARD, J.M., MEZZINA, M., and SARASIN, A.
(1995). Functional retroviral vector for gene therapy of xeroderma
pigmentosum group D patients. Hum. Gene Ther. 6, 1307–1315.
COLEMAN, A.B., and LUGO, T.G. (1998). Normal human
melanocytes that express a bFGF transgene still require exogenous
bFGF for growth in vitro. J. Invest. Dermatol. 110, 793–799.
D’ADDIO, M., PIZZIGONI, A., BASSI, M.T., BASCHIROTTO, C.,
VALETTI, C., INCERTI, B., CLEMENTI, M., DE LUCA, M., BAL-
LABIO, A., and SCHIAFFINO, M.V. (2000). Defective intracellu-
lar transport and processing of OA1 is a major cause of ocular al-
binism type 1. Hum. Mol. Genet. 9, 30113018.
DELLAMBRA, E., VAILLY, J., PELLEGRINI, G., BONDANZA, S.,
GOLISANO, O., MACCHIA, C., ZAMBRUNO, G., MENEGUZZI,
G., and DE LUCA, M. (1998). Corrective transduction of human epi-
dermal stem cells in laminin-5-dependent junctional epidermolysis
bullosa. Hum. Gene Ther. 9, 1359–1370.
DE LUCA, M., D’ANNA, F., BONDANZA, S., FRANZI, A.T., and
CANCEDDA, R. (1988). Human epithelial cells induce human me-
lanocyte growth in vitro but only skin keratinocytes regulate its
proper differentiation in the absence of dermis. J. Cell Biol. 107,
1919–1926.
DUNN, K.J., WILLIAMS, B.O., LI, Y., and PAVAN, W.J. (2000).
Neural crest-directed gene transfer demonstrates Wnt1 role in me-
lanocyte expansion and differentiation during mouse development.
Proc. Natl. Acad. Sci. U.S.A. 97, 10050–10055.
DUNN, K.J., INCAO, A., WATKINS-CHOW, D., LI, Y., and PAVAN,
W.J. (2001). In utero complementation of a neural crest-derived me-
lanocyte defect using cell directed gene transfer. Genesis 30, 70–76.
FERRERO, G.B., FRANCO, B., ROTH, E.J., FIRULLI, B.A., BOR-
SANI, G., DELMAS-MATA, J., WEISSENBACH, J., HALLEY, G.,
SCHLESSINGER, D., CHINAULT, A.C., ZOGHBI, H.Y., NEL-
SON, D.L., and BALLABIO, A. (1995). An integrated physical and
genetic map of a 35 Mb region on chromosome Xp22.3-Xp21.3.
Hum. Mol. Genet. 4, 1821–1827.
GALLICO, G.G., O’CONNOR, N.E., COMPTON, C.C., KEHINDE,
O., and GREEN, H. (1984). Permanent coverage of large burn
wounds with autologous cultured human epithelium. N. Engl. J. Med.
311, 448451.
GARNER, A., and JAY, B.S. (1980). Macromelanosomes in X-linked
ocular albinism. Histopathology 4, 243–254.
GUERRA, L., CAPURRO, S., MELCHI, F., PRIMAVERA, G., BON-
DANZA, S., CANCEDDA, R., LUCI, A., DE LUCA, M., and PEL-
LEGRINI, G. (2000). Treatment of stable” vitiligo by timed sur-
gery and transplantation of cultured epidermal autografts. Arch.
Dermatol. 136, 1380–1389.
HAMOEN, K.E., BOREL RINKES, I.H., and MORGAN, J.R. (2001).
Hepatocyte growth factor and melanoma: Gene transfer studies in
human melanocytes. Melanoma Res. 11, 89–97.
HEARING, V.J., and TSUKAMOTO, K. (1991). Enzymatic control of
pigmentation in mammals. FASEB J. 5, 2902–2909.
HIRSCHOWITZ, E.A., LEONARD, S., SONG, W., FERRIS, B.,
LEOPOLD, P.L., LEWIS, J.J., BOWNE, W.B., WANG, S.,
HOUGHTON, A.N., and CRYSTAL, R.G. (1998). Adenovirus-me-
diated expression of melanoma antigen gp75 as immunotherapy for
metastatic melanoma. Gene Ther. 5, 975–983.
IGOUCHEVA, O., and YOON, K. (2000). Targeted single-base cor-
rection by RNA–DNA oligonucleotides. Hum. Gene Ther. 11,
2307–2312.
INCERTI, B., CORTESE, K., PIZZIGONI, A., SURACE, E.M.,
VARANI, S., COPPOLA, M., JEFFERY, G., SEELIGER, M.,
JAISSLE, G., BENNETT, D.C., MARIGO, V., SCHIAFFINO,
M.V., TACCHETTI, C., and BALLABIO, A. (2000). OA1 knock-
out: New insights on the pathogenesis of ocular albinism type 1.
Hum. Mol. Genet. 9, 2781–2788.
KING, R.A., HEARING, V.J., CREEL, D.J., and OETTING, W.S.
(1995). Albinism. In The Metabolic and Molecular Bases of Inher-
ited Disease. C.R. Scriver, A.L. Beaudet, W.S. Sly, and D. Valle,
eds. (McGraw-Hill, New York) pp. 4353–4392.
KUCERA, G.T., BORTNER, D.M., and ROSENBERG, M.P. (1996).
Overexpression of an agouti cDNA in the skin of transgenic mice
recapitulates dominant coat color phenotypes of spontaneous mu-
tants. Dev. Biol. 173, 162–173.
MATHOR, M.B., FERRARI, G., DELLAMBRA, E., CILLI, M.,
MAVILIO, F., CANCEDDA, R., and DE LUCA, M. (1996). Clonal
analysis of stably transduced human epidermal stem cells in culture.
Proc. Natl. Acad. Sci. U.S.A. 93, 10371–10376.
MCKUSICK, V.A. (1998). Mendelian Inheritance in Man. Catalogs of
Human Genes and Genetic Disorders. 12
th
ed. (Johns Hopkins Uni-
versity Press, Baltimore).
MILLER, A.D., and ROSMAN, G.J. (1989). Improved retroviral vec-
tors for gene transfer and expression. BioTechniques 7, 980–986.
NESBIT, M., NESBIT, H.K.E., BENNETT, J., ANDL, T., HSU, M.Y.,
DEJESUS, E., MCBRIAN, M., GUPTA, A.R., ECK, S.L., and HER-
LYN, M. (1999). Basic fibroblast growth factor induces a trans-
formed phenotype in normal human melanocytes. Oncogene 18,
6469–6476.
O’DONNELL, F.E., HAMBRICK, G.W., GREEN, W.R., ILIFF, W.J.,
and STONE, D.L. (1976). X-linked ocular albinism: An oculocutaneous
macromelanosomal disorder. Arch. Ophthalmol. 94, 1883–1892.
PARK, K.C., CHINTAMANENI, C.D., HALABAN, R., WITKOP,
C.J., and KWON, B.S. (1993). Molecular analyses of a tyrosinase-
negative albino family. Am. J. Hum. Genet. 52, 406–413.
PELLEGRINI, G., TRAVERSO, C., FRANZI, A.T., ZINGIRIAN, M.,
CANCEDDA, R., and DE LUCA, M. (1997). Long term restoration
of damaged corneal surfaces with autologous cultivated corneal ep-
ithelium. Lancet 349, 990–993.
PELLEGRINI, G., RANNO, R., STRACUZZI, G., BONDANZA, S.,
GUERRA, L., ZAMBRUNO, G., MICALI, G., and DE LUCA, M.
(1999). The control of epidermal stem cells (holoclones) in the treat-
ment of massive full-thickness burns with autologous keratinocytes
cultured on fibrin. Transplantation 68, 868–879.
QUEVEDO, W.C., FITZPATRICK, T.B., SZABO, G., and JIMBOW,
K. (1987). Biology of melanocytes. In Dermatology in General Med-
icine. T.B. Fitzpatrick, A.Z. Eisen, K. Wolff, I.M. Freedberg, and
K.F. Austen, eds. (McGraw-Hill, New York) pp. 224251.
RAMA, P., BONINI, S., LAMBIASE, A., GOLISANO, O., PA-
TERNA, P., DE LUCA, M., and PELLEGRINI, G. (2001). Autolo-
gous fibrin-cultured limbal stem cells permanently restore the corneal
surface of patients with total limbal stem cell deficiency. Transplan-
tation 72, 1478–1485.
SCHIAFFINO, M.V., BASSI, M.T., GALLI, L., RENIERI, A., BRUT-
TINI, M., DE NIGRIS, F., BERGEN, A.A.B., CHARLES, S.J.,
YATES, J.R.W., MEINDL, A., LEWIS, R.A., KING, R.A., and
BALLABIO, A. (1995). Analysis of the OA1 gene reveals mutations
in only one-third of patients with X-linked ocular albinism. Hum.
Mol. Genet. 4, 23192325.
SCHIAFFINO, M.V., BASCHIROTTO, C., PELLEGRINI, G., MON-
TALTI, S., TACCHETTI, C., DE LUCA, M., and BALLABIO, A.
SCHIAFFINO ET AL.
956
(1996). The ocular albinism type 1 (OA1) gene product is a mem-
brane glycoprotein localized to melanosomes. Proc. Natl. Acad. Sci.
U.S.A. 93, 9055–9060.
SCHIAFFINO, M.V., D’ADDIO, M., ALLONI, A., BASCHIROTTO,
C., VALETTI, C., CORTESE, K., PURI, C., BASSI, M.T., COLLA,
C., DE LUCA, M., TACCHETTI, C., and BALLABIO, A. (1999).
Ocular albinism: Evidence for a defect in an intracellular signal trans-
duction system. Nat. Genet. 23, 108–112.
SCHNUR, R.E., GAO, M., WICK, P.A., KELLER, M., BENKE, P.J.,
EDWARDS, M.J., GRIX, A.W., HOCKEY, A., JUNG, J.H., KIDD,
K.K., KISTENMACHER, M., LEVIN, A.V., LEWIS, R.A.,
MUSARELLA, M.A., NOWAKOWSKI, R.W., ORLOW, S.J.,
PAGON, R.S., PILLERS, D.A., PUNNETT, H.H., QUINN, G.E.,
TEZCAN, K., WAGSTAFF, J., and WELEBER, R.G. (1998). OA1
mutations and deletions in X-linked ocular albinism. Am. J. Hum.
Genet. 62, 800–809.
SOBER, A.J., LEW, R.A., KOH, H.K., and BARNHILL, R.L. (1991).
Epidemiology of cutaneous melanoma. An update. Dermatol. Clin.
9, 617–629.
SVIDERSKAYA, E.V., BENNETT, D.C., HO, L., BAILIN, T., LEE,
S.T., and SPRITZ, R.A. (1997). Complementation of hypopigmen-
tation in p-mutant (pink-eyed dilution) mouse melanocytes by nor-
mal human P cDNA, and defective complementation by OCA2 mu-
tant sequences. J. Invest. Dermatol. 108, 30–34.
WONG, L., O’DONNELL, F.E., and GREEN, W.R. (1983). Giant pig-
ment granules in the retinal pigment epithelium of a fetus with X-
linked ocular albinism. Ophthalmic. Paediatr. Genet. 2, 47–65.
ZENG, L., QUILLIET, X., CHEVALLIER-LAGENTE, O., EVENO,
E., SARASIN, A., and MEZZINA, M. (1997). Retrovirus-mediated
gene transfer corrects DNA repair defect of xeroderma pigmentosum
cells of complementation groups A, B and C. Gene Ther. 4,
1077–1084.
ZHAO, H., BOISSY, Y.L., ABDEL-MALEK, Z., KING, R.A., NORD-
LUND, J.J., and BOISSY, R.E. (1994). On the analysis of the patho-
physiology of Chediak–Higashi syndrome. Defects expressed by cul-
tured melanocytes. Lab. Invest. 71, 25–34.
Address reprint requests to:
Dr. M. Vittoria Schiaffino
DIBIT, Scientific Institute San Raffaele
Via Olgettina 58
20132 Milan, Italy
E-mail:
schiaffino.mariavittoria@hsr.it
Received for publication July 5, 2001; accepted after revision
April 9, 2002.
Published online: April 23, 2002.
GENE TRANSFER IN HUMAN MELANOCYTES
957
... However, the process leading to macromelanosome formation and the actual role of the giant organelles in the pathogenesis of the disease remain unclear. In fact, the macromelanosomal phenotype displays variable expressivity in vitro and in vivo (34,35) and, at least in the mouse, manifests during development subsequent to optic misrouting, implying that it might represent only an epiphenomenon of the disease (27). In the search for the primary role of OA1 in ocular albinism, we identified an abnormally skewed distribution of melanosomes towards the apical pole of the RPE in Oa1-KO mice at embryonic stages preceding the formation of macromelanosomes and the reduction of organelle number. ...
... Constructs pR/OA1wt, pR/OA1T232K, LOA1SN and LOA1Δ18SN, containing the wild-type and mutant OA1 cDNAs, were previously described (22,35). Plasmids pEGFP-MC-LT and pEGFP-BR-LT were kindly provided by Dr J.A. Hammer III (31,32) and plasmid pEGFP-Rab27a was kindly provided by Dr M.C. ...
Article
Full-text available
The protein product of the ocular albinism type 1 gene, named OA1, is a pigment cell-specific G protein-coupled receptor exclusively localized to intracellular organelles, namely lysosomes and melanosomes. Loss of OA1 function leads to the formation of macromelanosomes, suggesting that this receptor is implicated in organelle biogenesis, however the mechanism involved in the pathogenesis of the disease remains obscure. We report here the identification of an unexpected abnormality in melanosome distribution both in retinal pigment epithelium (RPE) and skin melanocytes of Oa1-knock-out (KO) mice, consisting in a displacement of the organelles from the central cytoplasm towards the cell periphery. Despite their depletion from the microtubule (MT)-enriched perinuclear region, Oa1-KO melanosomes were able to aggregate at the centrosome upon disruption of the actin cytoskeleton or expression of a dominant-negative construct of myosin Va. Consistently, quantification of organelle transport in living cells revealed that Oa1-KO melanosomes displayed a severe reduction in MT-based motility; however, this defect was rescued to normal following inhibition of actin-dependent capture at the cell periphery. Together, these data point to a defective regulation of organelle transport in the absence of OA1 and imply that the cytoskeleton might represent a downstream effector of this receptor. Furthermore, our results enlighten a novel function for OA1 in pigment cells and suggest that ocular albinism type 1 might result from a different pathogenetic mechanism than previously thought, based on an organelle-autonomous signalling pathway implicated in the regulation of both membrane traffic and transport.
... Conversely, RPE cells have decreased pigmentation, likely because melanin is not evenly dispersed throughout the cell and is instead accumulated in a few giant melanosomes (Garner and Jay, 1980;Wong et al., 49 2009). GPR143 localizes to late endosomes/lysosomes and melanosomes, primarily to the ILVs (Basrur et al., 2003;Giordano et al., 2009;Samaraweera et al., 2001;Schiaffino et al., 1996;Schiaffino et al., 2002;Schiaffino and Tacchetti, 2005). Evidence suggests that GPR143 expression prevents fusion between multivesicular bodies and lysosomes when ectopically expressed in HeLa cells (Burgoyne et al., 2013); this suggests that in pigmented cells, GPR143 may be involved in segregating maturing melanosomes from lysosomes. ...
Article
Melanin synthesis is required for proper development and function of the visual system and for protection against ultraviolet radiation. Defects in melanin synthesis result in albinism, which is characterized by visual defects and increased skin cancer risk. Melanin is synthesized in pigment cells within specialized subcellular organelles called melanosomes. Some forms of albinism result from defects in melanosome maturation, but the underlying molecular mechanisms are incompletely understood. Melanosome maturation requires the trafficking of melanogenic cargoes to melanosome precursors and an increase in melanosome pH, thereby supporting activity of the enzyme tyrosinase to promote melanin synthesis. In this work, we investigate melanosome biogenesis and maturation in melanocytes derived from mouse models of two forms of albinism, (1) oculocutaneous albinism type 4 (OCA4) due to loss of function of SLC45A2, and (2) the Hermansky-Pudlak syndrome mouse model buff that has a missense mutation in VPS33A. (1) Here we show that SLC45A2, a putative sugar/proton symporter, localizes to melanosomes and increases organellar pH at its sites of localization. Further, we show that SLC45A2 likely functions at a later stage of melanosome maturation than the ion channel OCA2, which is also necessary to raise melanosomal pH and is defective in another subtype of OCA. Additionally, we show that a common SLC45A2-L374F variant associated with lighter pigmentation in humans is degraded more rapidly than the dark skin-associated L374 variant, indicating that decreased pigmentation reflects reduced proton export from melanosomes. (2) VPS33A, an SM protein that mediates fusion, is required in the endolysosomal and autophagosomal pathways, but whether it plays a direct role in melanosomal trafficking is unknown. Contrary to a previous report, we show that a VPS33A-D251E mutation in melan-bf cells does not prohibit pigmentation; instead, buff melanocytes cells contain enlarged, mature melanosomes despite partial mistrafficking of the mature melanosomal marker TYRP1. Replacement of wild-type VPS33A in wild-type melanocytes by VPS33A-D251E does not phenocopy buff melanocytes, suggesting that an additional mutation may be responsible for our observed buff phenotype. Our analyses of mouse albinism models have thus yielded significant insights into mechanisms of melanosome maturation.
... While transduction of primary human melanocytes by gamma-retroviral vectors has been reported [2], [3], [4], geneticin selection or co-culture with infected feeder cells, keratinocytes, or producer cell lines have been required to achieve high levels of transduction. Furthermore, retroviruses are unable to transduce non-dividing cells [5] and integration of the vector genome may lead to malignant transformation [6]. ...
Article
Full-text available
The study of melanocyte biology is important to understand their role in health and disease. However, current methods of gene transfer into melanocytes are limited by safety or efficacy. Recombinant adeno-associated virus (rAAV) has been extensively investigated as a gene therapy vector, is safe and is associated with persistent transgene expression without genome integration. There are twelve serotypes and many capsid variants of rAAV. However, a comparative study to determine which rAAV is most efficient at transducing primary human melanocytes has not been conducted. We therefore sought to determine the optimum rAAV variant for use in the in vitro transduction of primary human melanocytes, which could also be informative to future in vivo studies. We have screened eight variants of rAAV for their ability to transduce primary human melanocytes and identified rAAV6 as the optimal serotype, transducing 7-78% of cells. No increase in transduction was seen with rAAV6 tyrosine capsid mutants. The number of cells expressing the transgene peaked at 6-12 days post-infection, and transduced cells were still detectable at day 28. Therefore rAAV6 should be considered as a non-integrating vector for the transduction of primary human melanocytes.
... Melanoma research has been significantly hampered by the inability to transduce adequately genes of interest into primary human melanocytes. Previous investigators have attempted stable gene transfer into melanocytes using Moloney murine leukemia virusbased retroviral vectors with limited success (Mathor et al, 1996), whereas modified culture systems using feeder layers have increased the efficiency to 40% to 50% (Schiaffino et al, 2002). More recently, the use of adenoviral vectors for gene transfer into primary melanocytes has allowed for 90% to 100% transduction efficiency (Nesbit et al, 1999;McGill et al, 2002); however, these gene transduction systems can only allow for transient gene expression due to the inability to integrate efficiently into the host genome and are therefore not useful for long-term studies (reviewed in Ghazizadeh and Taichman, 2000). ...
Article
The Journal of Investigative Dermatology publishes basic and clinical research in cutaneous biology and skin disease.
... It encodes an orphan G-proteincoupled receptor, which crosses the melanosomal membrane. Oa1 is expressed exclusively in RPE and skin melanocytes [8][9][10][11][12] and its transcript is detectable in murine embryonic RPE from early stages of development [10]. A mouse knockout (KO) model, which shows some of the OA1 landmarks, has been generated [13]. ...
Article
Full-text available
X-linked recessive ocular albinism type I (OA1) is due to mutations in the OA1 gene (approved gene symbol GPR143), which is expressed in the retinal pigment epithelium (RPE). The Oa1 (Gpr143) knockout mouse (Oa1(-/-)) model recapitulates many of the OA1 retinal morphological anomalies, including a lower number of melanosomes of increased size in the RPE. The Oa1(-/-) mouse also displays some of the retinal developmental abnormalities observed in albino patients such as misrouting of the optic tracts. Here, we show that these anomalies are associated with retinal electrophysiological abnormalities, including significant decrease in a- and b-wave amplitude and delayed recovery of b-wave amplitude from photoreceptor desensitization following bright light exposure. This suggests that lack of Oa1 in the RPE impacts on photoreceptor activity. More interestingly, adeno-associated viral vector-mediated Oa1 gene transfer to the retina of the Oa1(-/-) mouse model results in significant recovery of its retinal functional abnormalities. In addition, Oa1 retinal gene transfer increases the number of melanosomes in the Oa1(-/-) mouse RPE. Our data show that gene transfer to the adult retina unexpectedly rescues both functional and morphological abnormalities in a retinal developmental disorder, opening novel potential therapeutic perspectives for this and other forms of albinism.
Article
Primary human melanocytes represent the precursor cells to melanoma and also specific targets of inherited and acquired pigmentation disorders [1-3]. We have developed a new gene transfer technology on primary adult human melanocytes by means of retroviral infection in absence of any toxic polycations currently employed to improve infection efficency (i.e. polybrene) that, as we show experimentally, induce apoptosis and DNA damage. We have cultured primary human melanocytes on a biocompatible nanostructured TiO2 film, obtained by the deposition of a supersonic beam of titania clusters [4-6], coated by retroviral vectors expressing GFP. By means of a "reverse infection" mechanism we achieved 80% of infection in absence of any toxic effect. We expect that such technology will allow efficient and safe genetic manipulation of primary cells for ex-vivo gene therapy.
Article
Little is known as to how cells ensure that organelle size and number are coordinated to correctly couple organelle biogenesis to the demands of proliferation or differentiation. OA1 is a melanosome-associated G-protein-coupled receptor involved in melanosome biogenesis during melanocyte differentiation. Cells lacking OA1 contain fewer, but larger, mature melanosomes. Here we show that OA1 loss-of-function reduces both the basal expression and the αMSH/cAMP-dependent induction of the microphthalmia-associated transcription factor (MITF), the master regulator of melanocyte differentiation. In turn, this leads to a significant reduction in expression of PMEL, a major melanosomal structural protein, but does not affect tyrosinase and melanin levels. In line with its pivotal role in sensing melanosome maturation, OA1 expression rescues melanosome biogenesis, activates MITF expression and thereby coordinates melanosome size and number, providing a quality-control mechanism for the organelle in which resides. Thus, resident sensor receptors can activate a transcriptional cascade to specifically promote organelle biogenesis. This article is protected by copyright. All rights reserved.
Article
Melanosomes are the specialized intracellular organelles of pigment cells devoted to the synthesis, storage and transport of melanin pigments, which are responsible for most visible pigmentation in mammals and other vertebrates. As a direct consequence, any genetic mutation resulting in alteration of melanosomal function, either because affecting pigment cell survival, migration and differentiation, or because interfering with melanosome biogenesis, transport and transfer to keratinocytes, is immediately translated into color variations of skin, fur, hair or eyes. Thus, over 100 genes and proteins have been identified as pigmentary determinants in mammals, providing us with a deep understanding of this biological system, which functions by using mechanisms and processes that have parallels in other tissues and organs. In particular, many genes implicated in melanosome biogenesis have been characterized, so that melanosomes represent an incredible source of information and a model for organelles belonging to the secretory pathway. Furthermore, the function of melanosomes can be associated with common physiological phenotypes, such as variation of pigmentation among individuals, and with rare pathological conditions, such as albinism, characterized by severe visual defects. Among the most relevant mechanisms operating in melanosome biogenesis are the signal transduction pathways mediated by two peculiar G protein-coupled receptors: the melanocortin-1 receptor (MC1R), involved in the fair skin/red hair phenotype and skin cancer; and OA1 (GPR143), whose loss-of-function results in X-linked ocular albinism. This review will focus on the most recent novelties regarding the functioning of these two receptors, by highlighting emerging signaling mechanisms and general implications for cell biology and pathology.
Article
Full-text available
Human keratinocytes isolated from a skin biopsy and cultured in vitro reconstitute a stratified squamous epithelium suitable for grafting on burned patients. Melanocytes coisolated from the same skin biopsy also proliferate under these culture conditions and maintain differentiated functions (i.e., synthesize melanin granules, regularly intersperse in the basal layer of the cultured epidermis, and transfer melanosomes in the cytoplasm of contiguous keratinocytes) (De Luca, M., A. T. Franzi, F. D'Anna, A. Zicca, E. Albanese, S. Bondanza, and R. Cancedda. 1988. Eur. J. Cell Biol. 46:176-180). Isolated melanocytes in culture grow in the presence of specific growth factors with a mean population doubling time of 4-10 d. In this paper we show that (a) human keratinocytes and oral epithelial cells possess strong and specific melanocyte growth stimulating activity (doubling time, 24 h); (b) melanocyte growth is not autonomous but requires close keratinocyte contact and is regulated to maintain a physiological melanocytes/keratinocytes ratiol and (c) pure skin keratinocytes, but not oral epithelial cells, have all the information required for the proper physiological location and differentiation of melanocytes in the epidermis.
Article
Full-text available
X-linked ocular albinism (OA1), Nettleship-Falls type, is characterized by decreased ocular pigmentation, foveal hypoplasia, nystagmus, photodysphoria, and reduced visual acuity. Affected males usually demonstrate melanin macroglobules on skin biopsy. We now report results of deletion and mutation screening of the full-length OA1 gene in 29 unrelated North American and Australian X-linked ocular albinism (OA) probands, including five with additional, nonocular phenotypic abnormalities (Schnur et al. 1994). We detected 13 intragenic gene deletions, including 3 of exon 1, 2 of exon 2, 2 of exon 4, and 6 others, which span exons 2-8. Eight new missense mutations were identified, which cluster within exons 1, 2, 3, and 6 in conserved and/or putative transmembrane domains of the protein. There was also a splice acceptor-site mutation, a nonsense mutation, a single base deletion, and a previously reported 17-bp exon 1 deletion. All patients with nonocular phenotypic abnormalities had detectable mutations. In summary, 26 (approximately 90%) of 29 probands had detectable alterations of OA1, thus confirming that OA1 is the major locus for X-linked OA.
Article
Ocular albinism type I (OA1) is an X-linked disorder characterized by severe reduction of visual acuity, strabismus, photophobia and nystagmus. Ophthalmologic examination reveals hypopigmentation of the retina, foveal hypoplasia and iris translucency. Microscopic examination of both retinal pigment epithelium (RPE) and skin melanocytes shows the presence of large pigment granules called giant melanosomes or macromelanosomes. In this study, we have generated and characterized Oa1-deficient mice by gene targeting (KO). The KO males are viable, fertile and phenotypically indistinguishable from the wild-type littermates. Ophthalmologic examination shows hypopigmentation of the ocular fundus in mutant animals compared with wild-type. Analysis of the retinofugal pathway reveals a reduction in the size of the uncrossed pathway, demonstrating a misrouting of the optic fibres at the chiasm, as observed in OA1 patients. Microscopic examination of the RPE shows the presence of giant melanosomes comparable with those described in OA1 patients. Ultrastructural analysis of the RPE cells, suggests that the giant melanosomes may form by abnormal growth of single melanosomes, rather than the fusion of several, shedding light on the pathogenesis of ocular albinism.
Article
Laminin-5 is composed of three distinct polypeptides, alpha3, beta3, and gamma2, which are encoded by three different genes, LAMA3, LAMB3, and LAMC2, respectively. We have isolated epidermal keratinocytes from a patient presenting with a lethal form of junctional epidermolysis bullosa characterized by a homozygous mutation of the LAMB3 gene, which led to complete absence of the beta3 polypeptide. In vitro, beta3-null keratinocytes were unable to synthesize laminin-5 and to assemble hemidesmosomes, maintained the impairment of their adhesive properties, and displayed a decrease of their colony-forming ability. A retroviral construct expressing a human beta3 cDNA was used to transduce primary beta3-null keratinocytes. Clonogenic beta3-null keratinocytes were transduced with an efficiency of 100%. Beta3-transduced keratinocytes were able to synthesize and secrete mature heterotrimeric laminin-5. Gene correction fully restored the keratinocyte adhesion machinery, including the capacity of proper hemidesmosomal assembly, and prevented the loss of the colony-forming ability, suggesting a direct link between adhesion to laminin-5 and keratinocyte proliferative capacity. Clonal analysis demonstrated that holoclones expressed the transgene permanently, suggesting stable correction of epidermal stem cells. Because cultured keratinocytes are used routinely to make autologous grafts for patients suffering from large skin or mucosal defects, the full phenotypic reversion of primary human epidermal stem cells defective for a structural protein opens new perspectives in the long-term treatment of genodermatoses.
Article
The retinal pigment epithelium (RPE) of a 21-week-old fetus with X-linked ocular albinism (XOA) was studied. Electron microscopy revealed giant pigment granules (macromelanosomes) with a vesiculoglobular substructure located in the equatorial and posterior regions. The giant pigment granules were similar to those found in the skin in neurofibromatosis, nevus spilus. xeroderma pigmentosum, and other conditions. Anteriorly, compound granules and melanosomes of unusual appearance that could be precursors of the macromelanosomes were present. Normal ellipsoidal melanosomes were observed in all areas, but were more common anteriorly.
Article
Background. Ocular burns cause depletion of limbal stem cells, which leads to corneal opacification and visual loss. Autologous cultured epithelial cells can restore damaged corneas, but this technology is still developing. We sought to establish a culture system that allows preservation of limbal stem cells and preparation of manageable epithelial sheets and to investigate whether such cultures can permanently restore total limbal stem cell deficiency. Methods. We selected a homogeneous group of patients whose limbal cell deficiency was evaluated by scoring the gravity of the clinical picture and the keratin expression pattern. Stem cells, obtained from the limbus of the contralateral eye, were cultivated onto a fibrin substrate and their preservation was evaluated by clonal analysis. Fibrin cultures were grafted onto damaged corneas. Results. Fibrin-cultured limbal stem cells were successful in 14 of 18 patients. Re-epithelialization occurred within the first week. Inflammation and vascularization regressed within the first 3-4 weeks. By the first month, the corneal surface was covered by a transparent, normal-looking epithelium. At 12-27 months follow-up, corneal surfaces were clinically and cytologically stable. Three patients had a penetrating keratoplasty approximately 1 year after restoration of their corneal surface. Their visual acuity improved from light perception or counting fingers to 0.8-1.0. Conclusions. Preservation of limbal stem cells in culture gives new perspectives on the treatment of ocular disorders characterized by complete limbal stem cell deficiency. The multicenter nature of this study and the handiness and ease of long-distance transportation of the fibrin-cultured epithelial sheets suggest that this technology can now be widely applied.
Article
Mutations in the P gene of humans and the homologous p-locus of mice, respectively, result in the homologous disorders oculocutaneous albinism type 2 (OCA2) and pink-eyed dilution. Although clearly required for melanin biosynthesis, the specific function of the P gene product, a melanosomal transmembrane protein expressed in melanocytes of the skin, hair and eyes, is not yet known. Here we describe lines of immortal melanocytes and melanoblasts from mice of the null genotype pcpp25H. These p-null melanocytes were severely hypopigmented, although they and the melanoblasts expressed mRNAs for a number of melanosomal proteins. Proliferation of the p-null melanoblasts was normal. Both diploid and immortal p-null melanocytes grew more slowly than wild-type melanocytes, however and were unusually susceptible to the antibiotic G418; these abnormalities were corrected by culture in high concentrations of L-tyrosine. Transfection of the p-null melanocytes with full-length normal human P cDNA resulted in complementation of deficient melanin biosynthesis and hypopigmentation. In contrast, transfection mutant human P cDNAs containing amino acid substitutions (A481T, V443I) found in patients with OCA2 resulted in minimal or partial correction, consistent with the corresponding pigmentation phenotypes in patients with these mutations. These results demonstrate the utility of this model system for distinguishing true OCA2 mutations from nonpathologic polymorphisms and for quantitating the effect of these mutations on P function.
Article
The generation of transgenic mice with mammalian genes cloned in yeast artificial chromosomes (YACs) has generated great interest in the field of gene transfer into livestock. Many of the problems associated with standard transgenesis—such as lack of crucial regulator elements and position effects related to the integration site, which lead to variation in expression levels irrespective of the dose of the transgene—have been practically overcome. The large size of YAC-derived gene constructs (in excess of 1 Mb) facilitates the presence and transfer of all elements required for the faithful regulation of a gene. With the experiments discussed in this report, we have addressed the possibility of applying the obvious advantages of YAC transgenesis to farm animals. We have generated transgenic rabbits carrying a 250 kb YAC covering the mouse tyrosinase gene by pronuclear microinjection, and thus rescued the albino phenotype of the transgenic individuals. To date, this is the first demonstration of a successful transfer of large genetic units into the germ line of farm animals. This development might improve the occurrence of transgene expression at physiological levels and specific sites in livestock. YAC transgenesis therefore will be applied in genetic engineering, for example, in the production of pharmacologically interesting proteins encoded by large gene units and generating transgenic donors for xenotransplantation. © 1996 Wiley-Liss, Inc.