Você está na página 1de 10

OVERVIEW OF BASIC SCIENCE SECTION 1

Skin Development and Maintenance 2 CHAPTER

Skin Development and Maintenance


Maranke I Koster, Cynthia A Loomis, Tamara Koss and David Chu

An overview of key events in the development of skin and its special-


Chapter Contents ized structures is shown in Figure 2.1.
Embryonic origin of the skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
Epidermal development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
EPIDERMAL DEVELOPMENT
Development of specialized cells within the epidermis . . . . . . . . . . . 57
The ectoderm that covers the developing embryo after gastrulation is
Development of the dermis and subcutis . . . . . . . . . . . . . . . . . . . . . . . . 58
a single-layered epithelium (Fig. 2.2A). The first step in epidermal
Development of the dermalepidermal junction . . . . . . . . . . . . . . . . . 59 development occurs when cells of the surface ectoderm adopt an epi-
Development of skin appendages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59 dermal fate1. Although this process does not result in major morpho-
logic changes, it is marked by dramatic alterations in gene expression
Skin stem cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
that result in the formation of the embryonic epidermis, which initially
Prenatal diagnosis of genodermatoses . . . . . . . . . . . . . . . . . . . . . . . . . . 63 consists of only a basal layer (Fig. 2.2B). Basal keratinocytes subse-
Significance of skin development in postnatal life . . . . . . . . . . . . . . . 63 quently generate cells of the periderm, a single cell layer that covers the
developing epidermis until the cornified cell layer is formed (Figs
2.2CF & 2.3A,B). Although the precise function of the periderm is not
known, it is believed to participate in the exchange of substances across
INTRODUCTION fetal skin and protects the developing epidermis from the amniotic
fluid.
Development of the human embryo is a complex process involving The embryonic epidermis begins to stratify at approximately 8 weeks
highly orchestrated cell movements, proliferation, death and differen- estimated gestational age (EGA)2. At this time, basic organogenesis is
tiation. This chapter focuses on key events and regulatory mechanisms complete and bone marrow hematopoiesis commences, marking the
that result in skin morphogenesis, maintenance and regeneration. The transition from embryo to fetus. During the first stage of epidermal
spectrum of cutaneous abnormalities that can result from mutations stratification, an intermediate cell layer is formed between the basal
in genes with critical roles in skin development is discussed. Ulti- layer and periderm (Figs 2.2D & 2.3B). Unlike suprabasal keratinocytes
mately, improved understanding of the pathways and signaling cascades in the postnatal epidermis, the intermediate layer consists of actively
that are disrupted in genetic skin disorders will aid in the development proliferating cells. As a consequence, it is able to expand to accom-
of therapeutic approaches for patients with acquired as well as inherited modate the rapid growth of the embryo as well as create additional
skin disorders. layers of intermediate cells over the next several weeks (Fig. 2.2E).
However, the intermediate cell layer only exists temporarily during
epidermal development, as it is eventually replaced by layers of post-
EMBRYONIC ORIGIN OF THE SKIN mitotic keratinocytes undergoing terminal differentiation.
The skin is composed of diverse cell types of both ectodermal (e.g. Terminal differentiation, the process resulting in the formation of
keratinocytes, melanocytes, Merkel cells, neurons) and mesodermal mature keratinizing epidermal cells, begins during the second trimes-
(e.g. fibroblasts, hematopoietic cells such as Langerhans cells, endothe- ter. Early cornification can be observed within the hair canal at
lial cells) lineages. To understand the origins of these cells, it is impor- approximately 15 weeks EGA2a, but it does not commence in the
tant to review the early stages of embryogenesis. Immediately after interfollicular epidermis until 2224 weeks EGA, occurring first in
fertilization, cells divide rapidly, and by the end of the first week, the the skin on the head, palms and soles. Expression of the p63 gene is
embryo begins to implant into the uterine wall. During the third week, required for the process of epidermal stratification. The process begins
the embryo undergoes gastrulation, a complex process resulting in the when cells in the intermediate layer permanently withdraw from the
formation of the three embryonic germ layers: endoderm, mesoderm cell cycle and differentiate into spinous and granular cells (Fig. 2.2F).
and ectoderm. The cornified cell layer, which is composed of dead keratinocytes
During the next stage of embryogenesis, ectodermal cells commit to (corneocytes) held together by a matrix of proteins and lipids (see Ch.
either a surface ectodermal or neuroectodermal fate. The surface ecto- 56), subsequently starts to form and is several cells thick by 2426
dermal cells eventually differentiate into the keratinocytes of the weeks EGA. The corneocytes are a reflection of the closely regulated
embryonic epidermis, whereas the neuroectodermal cells invaginate to process of terminal differentiation that is required for normal func-
create the neural tube in a process called neurulation. As the neural tioning of the skin. At the same time, the periderm detaches from the
tube forms, cells in its dorsal portion separate to form the neural crest. underlying epidermis and is sloughed off into the amniotic fluid, with
An important neural crest-derived cell in the skin is the melanocyte. remnants contributing to the vernix caseosa that coats newborns.
Although Merkel cells were once believed to be neural crest derivatives, During the third trimester, the number of keratohyalin and lamellar
it was recently established that they descend from the epidermal granules as well as stratum corneum layers increase. By the mid third
lineage. The lineage of the dermis depends on its location. Whereas the trimester, the epidermis is morphologically similar to adult skin (Figs
dermis (and other mesenchymal structures) of the face and frontal scalp 2.2G & 2.3C), although it does not acquire full barrier function until
are derived from the neural crest, the dermis elsewhere is derived from a few weeks after birth.
the mesoderm. Knowing from which germ layer and lineage different
cell types in the skin derive helps one to understand the pathophysiol- Clinical Relevance
ogy of cutaneous disorders ranging from congenital melanocytic nevi Genetic abnormalities affecting various stages of epidermal morpho-
(where neurotization is explained by the derivation of both melano- genesis have been found to underlie inherited skin disorders in
cytes and peripheral nerve sheath cells from the neural crest) to humans. However, generalized abnormalities in epidermal specifica-
Waardenburg syndrome (where craniofacial dysmorphism and hearing tion, the process through which the surface ectoderm adopts an epi-
impairment as well as pigmentary abnormalities reflect disrupted dermal fate, have not been identified, and it is likely that such defects 55
migration and survival of neural crest-derived cells). would be incompatible with survival past the first trimester. Mosaic
Fig. 2.1 Critical events in the development of the
CRITICAL EVENTS IN THE DEVELOPMENT OF SKIN AND ITS SPECIALIZED STRUCTURES skin and its specialized structures. The timeline
indicates the time of initiation defined by estimated
gestational age (EGA) and duration of pregnancy (by
Clinical timing Fetal age Epidermal Dermal-subcutis Appendage last menstrual period [LMP]). Refers to skin on the
SECTION
of pregnancy development development development
1 LMP
(weeks)
EGA
(weeks)
back unless otherwise noted.
Overview of Basic Science

6 4
Periderm
Basal layer Tooth primordia
Periderm Nail primordia
10 8 Intermediate layer Volar eccrine gland
Dermal-subcutaneous
Basal layer boundary distinct primordia
Melanocytes Hair follicle/sebaceous
Langerhans cells gland primordia
14 12
Merkel cells

Papillary and
18 16
reticular dermal
boundary distinct
Second trimester

22 20 Dermal ridges
Trunk eccrine
gland primordia
Periderm sloughs
26 24
Stratum corneum
Granular layer
Spinous layer
30 28 Basal layer

34 32

38 36

42 40 Birth

3 weeks'
postnatal age

Fig. 2.2 Development of the epidermis. A The


DEVELOPMENT OF THE EPIDERMIS epidermis develops from the surface ectoderm,
a single-layered epithelium that initially covers the
developing embryo. B Through changes in gene
A B expression, cells of the surface ectoderm adopt an
epidermal fate, resulting in the formation of the basal
layer of the epidermis. C Basal cells subsequently
give rise to the periderm, a cell layer that covers the
C D
developing epidermis until cornification occurs.
D Epidermal stratification begins with the formation
of a highly proliferative intermediate layer between
the basal layer and the periderm. E The intermediate
layer becomes several cells thick over the next few
weeks. F Intermediate cells ultimately withdraw from
the cell cycle and differentiate into spinous and
granular keratinocytes. G The periderm is replaced by
E F
the cornified cell layer.

G Surface ectoderm Spinous layer

Basal layer Granular layer

Periderm Cornified layer

Intermediate layer
56
Fig. 2.3 Development of the
skin. A At 36 days estimated
gestational age, the embryonic
epidermis consists of two layers
a superficial periderm layer CHAPTER

A
overlying a basal layer. B By 72
days, the basal layer has given
rise to the highly proliferative
2

Skin Development and Maintenance


intermediate cell layer. C A
distinct granular cell layer and
stratum corneum are seen in
neonatal skin. D An early
peg-stage follicle from a mid
second trimester fetus. The
more superficial bulge consists
of the developing sebaceous
gland (arrowhead), whereas the
deeper bulge represents the
B
insertion point of the future
arrector pili muscle and the
location of the presumptive
follicular stem cells (arrow).
Photomicrographs courtesy of Dr Karen
Holbrook, from Loomis CA, Koss T, Chu D.
Fetal skin development. In: Eichenfield LF,
Frieden IJ, Esterly NB (eds). Textbook of
Neonatal Dermatology, 2nd edn.
Philadelphia: WB Saunders, 2007:117.

C D

skin conditions that result in abnormalities of the epidermis (e.g. of infection, dehydration, and excessive absorption of topical medications
keratinocytes or melanocytes) and/or its appendages often have a dis- or chemicals4. Even healthy full-term infants do not attain full skin
tribution pattern that follows Blaschkos lines, which are thought to barrier function until 3 weeks of age. The structural features of prema-
represent the migration pathways of epidermal cells during embryonic ture skin and adult skin are summarized in Table 2.1.
development. Classic examples include epidermal nevi (with underly-
ing defects including mosaicism for mutations in genes encoding
the fibroblast growth factor receptor 3, keratin 1 or keratin 10) and DEVELOPMENT OF SPECIALIZED CELLS
sebaceous nevi (where the underlying genetic defect remains unknown WITHIN THE EPIDERMIS
but is thought to be lethal if present in all the cells of the body) (see
Ch. 62). Two populations of specialized cells melanocytes and Langerhans cells
There are a number of genetic disorders that result in abnormal migrate to the epidermis during early embryonic development.
epidermal differentiation and barrier formation. One clinical presenta- Melanocytes are derived from the neural crest that forms along the
tion of such conditions is a collodion baby born encased in a taut, dorsal neural tube. Melanocyte precursors migrate away from the
shiny, transparent membrane that is formed by aberrant stratum neural tube within the mesenchyme beneath the primitive epidermis.
corneum. After shedding the membrane, most of these infants manifest They follow a characteristic trajectory, moving dorsolaterally and then
with lamellar ichthyosis (LI) or congenital ichthyosiform erythroderma ventrally around the trunk to the ventral midline, anteriorly over the
(CIE), two forms of autosomal recessive ichthyosis that exist on a scalp and face, and distally along the extremities. Cutaneous melano-
spectrum3. However, the phenotypes that can develop following a collo cytes can also arise from Schwann cell precursors located along nerves
dion membrane include not only the large, plate-like brown scales of in the skin, which originate from the neural crest via the distinct
LI and the erythroderma with fine white scales of CIE, but also com- ventral pathway5.
pletely normal appearing skin. Referred to as a self-healing collodion Melanocytes are present in the epidermis by the middle of the first
baby, the latter is an example of a dynamic epidermal phenotype that trimester, but they are not fully functional until the second trimester.
depends on environmental conditions. All of these outcomes can result Melanocytes can be first identified within the embryos epidermis at
from mutations in the same set of genes that encode proteins essential approximately 50 days EGA, based on their positive reactivity with the
for formation of the epidermal barrier, including transglutaminase-1 HMB45 monoclonal antibody and their dendritic morphology. Epider-
(an enzyme that cross-links lipids to the cornified cell envelope; TGM1), mal melanocyte density is high early in embryonic development (1000
lipid processing enzymes (ALOXE3, ALOX12B) and lipid transporters cells/mm2), and it increases further (~3000 cells/mm2) as the epidermis
(ABCA12) (see Ch. 57). stratifies (8090 days EGA) and the appendages begin to develop; later
More deleterious mutations in the ABCA12 gene represent the cause in gestation, the density decreases and becomes similar to that of young
of harlequin ichthyosis (HI), an especially severe and often fatal disorder adults (8001500 cells/mm2). However, epidermal melanin production
of cornification characterized by aberrant epidermal maturation. does not begin until 34 months EGA, and melanosome transfer to
Patients with HI are born with a tremendously thick, armor-like shell keratinocytes is not seen until 5 months EGA. Even though all melano-
of hyperkeratosis, severe ectropion and eclabium, and underdevelop- cytes are functional and in place at birth, newborn skin is not fully
ment of the nose and ears. The extreme phenotype of HI highlights the pigmented and subsequently darkens over the first few months
importance of lipid transport into lamellar bodies for epidermal forma- of life; this process is most pronounced in more darkly pigmented
tion and function. infants.
Abnormalities in the stratum corneum are present not only in infants Active melanocytes are also present throughout the dermis during
with ichthyosis, but also in premature infants, especially those born embryonic development. Eventually, most of these dermal melanocytes
before 28 weeks EGA. The immaturity of the stratum corneum results migrate to the epidermis or undergo apoptosis. By the time of birth, 57
in impaired barrier function, which leads to an increased risk of dermal melanocytes have disappeared, with the exception of certain
COMPARATIVE FEATURES OF PREMATURE, NEWBORN, AND ADULT SKIN

Premature Newborn Adult


SECTION
Skin thickness 0.9mm 1.2mm 2.1mm
1 Epidermal surface Vernix (gelatinous) Vernix Dry
Epidermal thickness ~2025microns ~4050microns ~50microns
Overview of Basic Science

Stratum corneum 45microns 910microns 915microns


thickness
56 cell layers >15 cell layers >15 cell layers
Spinous cell glycogen Abundant Little or none Little or none
content
Melanocytes High number of cells; few mature Similar number of cells to young Numbers decrease with age; melanin production
melanosomes adult; low melanin production dependent on skin type, body area
Dermalepidermal All known adult antigens expressed; Structural features and antigens Well-developed adhesive structures; large
junction fewer and smaller desmosomes similar to those of the adult number of antigens expressed
Papillary dermis
Boundary with reticular Present but not marked Present but not marked Marked
dermis
Size of collagen fiber Small Small Small
bundles
Cellular density Abundant Abundant Moderately abundant
Reticular dermis
Boundary with the Marked Marked Marked
subcutis
Size of collagen fiber Small Small to intermediate Large
bundles
Cellular density Abundant Moderately abundant Sparse
Elastic fibers Sparse; tiny with immature Small size and immature structure; Large in reticular dermis, small and immature in
structure distribution similar to adult papillary dermis; form network
Hypodermis Well-developed fatty layer Well-developed fatty layer Well-developed fatty layer
Table 2.1 Comparative features of premature, newborn, and adult skin. Reproduced with permission from Schachner LA, Hansen RC (eds). Pediatric Dermatology, 4th edn. London: Mosby, 2010.

anatomic sites (head and neck, dorsal aspects of the distal extremities have been identified, including those encoding transcription factors
and presacral area), which correspond to the most common locations (e.g. microphthalmia-associated transcription factor [MITF], PAX3,
for dermal melanocytoses and blue nevi (see Ch. 112). SOX10, SNAI2) as well as membrane receptors and their ligands (e.g.
Langerhans cell precursors appear within the epidermis during the endothelin-3, endothelin B receptor, KIT receptor)8 (see Ch. 66). The
first trimester and are first detectable in this site as early as 40 days endothelin B receptor is found on melanoblastganglion cell precursors
EGA. These cells can be distinguished by their characteristic dendritic in the developing neural crest, explaining the Hirschsprung disease (as
morphology; expression of CD45, HLA-DR and CD1c; and high levels well as pigmentary abnormalities) that is associated with defects in this
of ATPase activity. Langerin expression precedes CD1a acquisition, receptor or its ligand.
which first occurs at 13 weeks EGA, and the production of Birbeck
granules6. The density of Langerhans cells in fetal skin remains low
early in gestation and increases to typical adult levels during the third DEVELOPMENT OF THE DERMIS AND SUBCUTIS
trimester. The specification of dermal mesenchymal cells is a complex process
Merkel cells, highly innervated neuroendocrine cells involved in that is incompletely understood. Unlike the epidermis, which is
mechanoreception, are initially identified within the epidermis during derived exclusively from the ectoderm, the origin of the dermis varies
the first trimester. These cells are detected as early as 812 weeks EGA depending on the body site. As noted above, the dermal mesenchyme
in palmoplantar epidermis, and slightly later in interfollicular of the face and anterior scalp (as well as the underlying muscle and
skin. Merkel cells are identified by the presence of cytoplasmic dense bone) is derived from neural crest ectoderm, thereby explaining the
core granules, cytokeratin 20 and neuropeptides. They are found in facial dysmorphia in the neurocristopathy Waardenburg syndrome.
the basal layer of the epidermis, are often associated with appendages On the other hand, the dermal mesenchyme of the back originates
and nerve fibers, and are particularly dense on volar skin. The devel- from the dermomyotome of the embryonic somite, and the dermal
opmental origin of Merkel cells has been the subject of longstanding mesenchyme of the extremities and ventral trunk is thought to arise
controversy, but recent studies have demonstrated that mammalian from the lateral plate mesoderm.
Merkel cells are derived from an epidermal rather than neural crest By 68 weeks EGA, presumptive dermal fibroblasts are situated
lineage7. under the developing epidermis9. However, at this developmental stage,
there is no distinct demarcation between the cells that will give rise to
Clinical Relevance the dermis and those that will give rise to musculoskeletal components.
Several inherited pigmentary disorders result from genetic defects that Although embryonic dermal cells are capable of synthesizing collagens
lead to abnormal migration and proliferation of neural crest-derived (e.g. types I, III and IV) and some microfibrillar components, these
melanocyte precursors (melanoblasts). Piebaldism and Waardenburg proteins are not yet assembled into complex fibers. Of note, the ratio
syndrome are characterized by achromic patches of skin on the central of collagen III to collagen I is 3:1, the reverse of what is seen in the
forehead, central abdomen and extremities, a distribution pattern adult dermis.
that reflects the failure of melanocyte precursors to survive, proliferate The demarcation between the dermis and underlying skeletal con-
58 or travel to the distal points of their embryonic migration pathway. densations becomes distinct at approximately 9 weeks EGA. By 1215
A number of different causative genes leading to this phenotype weeks EGA, progressive changes in matrix organization and cell
morphology distinguish the fine weave of the papillary dermis located embryonic DEJ acquires the rete ridges and dermal papillae that char-
directly beneath the epidermis from the thicker, deeper reticular dermis. acterize the adult DEJ.
At this stage, the collagen proteins produced by the fibroblasts start to
assemble into collagen fibers, which continue to accumulate in the Clinical Relevance CHAPTER
reticular dermis during the second and third trimesters. Electron micro-
scopy can first detect definitive elastic fibers at approximately 2224
weeks EGA. As development progresses, the watery, proteoglycan-rich,
Epidermolysis bullosa (EB) is a heterogeneous group of genetic disorders
characterized by blister formation due to mechanical fragility of the 2
cellular dermis of the embryo is modified to the more rigid, fibrous, skin. EB can result from mutations in genes that encode several

Skin Development and Maintenance


acellular dermis characteristic of adult skin. By the end of the second different components of the DEJ12. The specific protein affected (and
trimester, the dermis shifts from a non-scarring to a scarring form of the degree to which it is altered) largely determines disease severity,
wound repair. At birth, the dermis is still thinner and more cellular depth of bulla formation, and involvement of extracutaneous tissues
than the adult dermis. (see Ch. 32).
The basic pattern of the dermal vasculature is discernible by the end
of the first trimester. This pattern, however, undergoes extensive
remodeling in utero and is not fully mature until after birth. Numerous DEVELOPMENT OF SKIN APPENDAGES
molecular controls are involved in the regulation of angiogenesis, Skin appendages (hair, nails, and eccrine, apocrine and sebaceous
including the vascular endothelial growth factor (VEGF) family as well glands) have both epidermal and dermal components that are critical
as the tyrosine kinase receptors Tie-1 and Tie-2 and their stimulatory in embryogenesis. Normal development of skin appendages depends on
(Ang-1) and inhibitory (Ang-2) angiopoietin ligands (see Ch. 102). tightly regulated interactions between the early dermis and epidermis
Nerve networks are formed by the mid to late first trimester, and (e.g. signaling molecules that are sent back and forth between these
they tend to follow the vascular pattern. Accumulation of subcutane- tissues), and a failure in either component (or in communication
ous fat begins during the second trimester and continues through the between them) leads to aberrant development. Despite the differences
third trimester, when distinct lobules separated by fibrous septae are in the structure and function of the various types of cutaneous append-
formed. ages, the developmental processes that regulate their formation are
remarkably similar13.
Clinical Relevance
Mutations in the genes that encode a variety of dermal structural pro-
teins (e.g. collagens, components of elastic fibers) and the enzymes that
Hair Follicle Development
process them underlie multiple forms of EhlersDanlos syndrome The complex dermalepidermal interactions and genetic control mech-
(characterized by skin hyperextensibility and fragility with poor wound anisms that occur in appendage development are best understood in
healing) and cutis laxa (featuring lax, redundant skin). Genetic defects the formation of the hair follicle. Follicle formation is initiated by
in regulatory proteins important for extracellular matrix development signals from the dermis that direct the embryonic epidermis to form
can lead to abnormalities in the skin and other organs. For example, focal thickenings, called placodes (see Figs 68.2 & 68.3)14. Placodes are
mutations in the latent transforming growth factor- (TGF-) binding first seen on the scalp and face between 10 and 11 weeks EGA; they
protein-4 gene (LTBP4) were recently found to underlie a form of cutis subsequently develop in a caudal and then in a ventral direction. The
laxa associated with severe pulmonary, gastrointestinal and genitourin epidermal placodes instruct the underlying dermal cells to condense
ary malformations. The absence of LTBP4, an extracellular matrix and form the presumptive dermal papilla. The dermal papilla then
protein that binds to fibrillin and controls the bioavailability of TGF-, directs the keratinocytes of the placode to proliferate and extend deeper
leads to aberrant TGF- activity and defective elastic fiber assembly in into the dermis, thereby forming the hair germ (see Figs 68.2 & 68.3).
many types of tissues. By 1214 weeks EGA, the base of the developing hair follicle surrounds
Another disorder characterized by abnormal dermal development is the presumptive dermal papilla, forming the hair peg (see Figs 68.2 &
Goltz syndrome (focal dermal hypoplasia), an X-linked dominant dis- 68.3). The superficial portion of the developing hair follicle has two
order characterized by areas of dermal hypoplasia (with fat herniation/ distinct bulges. The more superficial bulge consists of the developing
hamartomas) in a distribution following Blaschkos lines, skeletal sebaceous gland, whereas the deeper bulge represents the insertion
defects and mucosal papillomas. Goltz syndrome is caused by muta- point of the future arrector pili muscle and the location of the presump-
tions in PORCN, an effector of the Wnt signaling pathway that is tive follicular stem cells.
critical for normal dermal development10. Blaschkos lines are thought Hair follicles undergo further maturation during the second trimester,
to represent pathways of epidermal development, and in a mouse forming seven concentric cell layers (from outer to inner): outer root
model, PORCN expression and resultant Wnt signaling originating sheath; inner root sheath, composed of Henleys and Huxleys layers
from the ectoderm have been shown to regulate development of the and a cuticle; and then the hair shaft cuticle, cortex and medulla (see
dermis. Fig. 68.2). The hair canal is fully formed by 1821 weeks EGA, and
hairs are visible shortly thereafter. The hairs continue to grow until
2428 weeks EGA, when they leave the active growing phase (anagen)
DEVELOPMENT OF THE DERMAL and enter the short-lasting degenerative phase (catagen) and subse-
EPIDERMAL JUNCTION quently the resting phase (telogen). The follicles then start the second
hair cycle by re-entering anagen; new hairs grow and push out the first
The dermalepidermal junction (DEJ) mediates adhesion between the group of telogen hairs, which are shed into the amniotic fluid.
basal keratinocytes and the dermis and provides resistance against Hair cycling through anagen, catagen and telogen continues through-
shearing forces on the skin (see Ch. 28). The DEJ develops from a out life, but the hair cycles for individual hair follicles become asyn-
simple, generic basement membrane in the embryo to a highly complex, chronous after birth. The third hair cycle is initiated perinatally, leading
multilayered structure in the second-trimester fetus. to the shedding of the second wave of fine lanugo hair follicles. Most
The embryonic DEJ consists of a lamina densa and a lamina lucida, hairs become thicker and coarser with subsequent growth cycles,
and it is composed of molecules that are common to all basement leading to vellus and then adult-like terminal hairs on the scalp and in
membrane zones (e.g. type IV collagen, laminin, heparan sulfate, pro- the eyebrows.
teoglycans)11. The DEJ becomes progressively more complex, and skin- Many genes important for hair follicle development and cycling have
specific components of the DEJ start to appear after the embryonicfetal been identified (see Ch. 68). Sonic hedgehog (SHH), a signaling mole-
transition at 8 weeks EGA, coincident with the onset of epidermal cule secreted by cells of the developing hair follicle, is required for the
stratification. By 12 weeks EGA, almost all of the structures charac- maturation of the dermal papilla and for the progression of the follicle
teristic of the mature DEJ are in place (see Ch. 28). Hemidesmosomes, placode to the peg hair stage. SHH also appears to be critical in mediat-
anchoring filaments and anchoring fibrils are synthesized by basal ing the transition from telogen to anagen during postnatal hair cycling.
keratinocytes, with the type VII collagen-containing anchoring fibrils Members of the Wnt, bone morphogenetic protein (BMP) and fibroblast
localizing to the sub-lamina densa; laminin 5 and the bullous pemphi- growth factor (FGF) families of signaling molecules are also important 59
goid antigens are also expressed. As development progresses, the flat in hair follicle development and cycling. Transcription factors with roles
in follicular differentiation have also been identified, including HOXC13 relatively common type of ectodermal dysplasia that affects the sweat
and FOXN1. glands (hypohidrosis), hair (hypotrichosis) and teeth (hypodontia). HED
Sebaceous gland development parallels follicular development. The is caused by mutations in genes encoding components of the ectodys-
presumptive sebaceous gland is first seen at around 1316 weeks EGA plasin A (EDA) signaling pathway, which is critical for the initiation of
SECTION
as the most superficial bulge on the developing hair follicle. The outer sweat gland, hair follicle and tooth morphogenesis. Underlying muta-
1 proliferative layer of the sebaceous gland generates lipogenic cells that
progressively accumulate lipid/sebum until they are terminally differ-
tions can occur in the genes encoding the EDA ligand, EDA receptor
(EDAR), or EDAR-associated death domain (an intracellular adaptor
entiated, at which time they disintegrate and release their products into protein that functions in EDAR signaling). Mutations in EDA are
Overview of Basic Science

the upper portion of the hair canal. Maternal hormones contribute to responsible for the X-linked variety of HED (XLHED), which leads to
sebaceous gland hypertrophy and increased synthesis and secretion of full-blown disease in affected males and areas of involved skin fol-
sebum during the second and third trimesters. The neonatal adrenal lowing Blaschkos lines in heterozygous females. Because the underly-
gland also has a disproportionately large zone of androgen production ing defect is in a soluble ligand that is only required for a brief period
(referred to as the fetal zone), which involutes over the first year during the initiation of appendage formation, XLHED is a prime target
of life. for protein therapy. Encouragingly, studies in mice and dogs have dem-
onstrated an almost complete correction of the XLHED phenotype
Nail Development upon prenatal or neonatal treatment with recombinant EDA protein16,17.
Nail development begins at 810 weeks EGA and is completed by the Clinical trials with recombinant EDA protein for the treatment of
fifth month of intrauterine development. The flat, rectangular surface infants with XLHED are planned.
of the future nail bed on the dorsal digital tips is first demarcated by In addition to mutations in genes that are required for appendage
folds visible at 810 weeks EGA. A wedge of ectoderm invaginates development, ectodermal dysplasias can also be caused by mutations
obliquely into the mesenchyme along the proximal end of the early nail in genes that are required for epidermal development18. In these
field, forming the proximal nail fold by 13 weeks EGA. The presump- instances, abnormal appendages result from disruption of cross-talk
tive nail matrix cells, which will later produce the differentiated nail between the developing epidermis and dermis. Mutations in p63, a gene
plate, are found ventral to the proximal nail fold. The nail bed on the that is required for epidermal morphogenesis, can cause several types
dorsal digit is the first skin structure to keratinize, at around 11 weeks; of ectodermal dysplasia, including the ankyloblepharonectodermal
keratinization begins distally and then continues over the nail bed dysplasiacleft lip/palate (AEC) and ectrodactylyectodermal dysplasia
toward the proximal nail fold. The first, preliminary nail is shed easily cleft lip/palate (EEC) syndromes (see Ch. 63). The phenotypic differ-
and replaced by the hard, differentiated nail plate that emerges from ences between AEC and EEC are explained by underlying mutations in
under the proximal nail fold during the fourth month of gestation and separate regions of the p63 gene, which affect different functions and
completely covers the nail bed by the fifth month. isotypes of the p63 protein.
Wnt signaling is critical for the development of epidermal append-
Eccrine and Apocrine Sweat Gland Development ages, and genetic defects in this pathway underlie conditions ranging
from hereditary hypotrichosis simplex to onycho-odonto-dermal dys-
Like hair and nails, palmoplantar eccrine sweat glands begin to develop plasia and SchpfSchulzPassarge syndrome19. The nevoid basal cell
during the first trimester and are fully developed by the second trimes- carcinoma syndrome (Gorlin syndrome) is caused by mutations in the
ter. Sweat gland development starts with the formation of large mes- PTCH tumor suppressor gene that result in overactivation of the sonic
enchymal bulges or pads (analogous to the paw pads of other mammals) hedgehog (SHH) signaling pathway, which has important roles in neu-
on the palms and soles between 55 and 65 days EGA. Parallel ectoder- rologic and anteriorposterior axis development as well as hair follicle
mal ridges are induced in the epidermis overlying these pads between formation20. This explains the broad range of developmental anomalies,
12 and 14 weeks EGA. The curves and whorls formed by these ridges including craniofacial and skeletal defects, and the propensity for
result in characteristic dermatoglyphics (fingerprints), which can be medulloblastomas, odontogenic keratocysts and multiple basal cell car-
seen on the digit tips by the fifth month of gestation. Unlike most other cinomas to arise (see Ch. 108).
animals, the mesenchymal pads in the human fetus regress by the third
trimester.
Beginning between 14 and 16 weeks EGA, individual eccrine gland SKIN STEM CELLS
primordia bud along the ectodermal ridges at regularly spaced intervals.
The buds elongate as cords of cells that enter the pad mesenchyme. By Stem cells are present in each self-renewing tissue and are responsible
16 weeks EGA, glandular structures form at the terminal portion for maintaining and repairing the tissue they reside in. The epidermis,
of the buds and secretory and myoepithelial cells are visible. Canaliza- dermis, appendages, and melanocytes are each maintained by different
tion of the dermal component of the eccrine duct is also complete by stem cell compartments. Stem cells are self-renewing and divide
16 weeks EGA; this occurs via a loss of the desmosomal adhesions infrequently during periods of homeostasis, with the latter feature
along the innermost ectodermal surfaces, with concomitant mainte- believed to protect them from acquiring mutations during cell cycle
nance of the adhesion between duct cells and the gland walls. Canaliza- progression. Under homeostatic conditions, stem cells are believed to
tion of the epidermal component of the duct is not complete until 22 undergo asymmetric division, generating one new stem cell and one
weeks EGA. daughter transit amplifying (TA) cell. Unlike stem cells, TA cells divide
Unlike volar eccrine sweat glands, interfollicular eccrine glands and frequently and ultimately differentiate into lineage-specific cell types.
apocrine glands do not begin to form until the fifth month of gestation. Within the skin, stem cells that maintain the epidermis and hair fol-
Like sebaceous glands, apocrine glands typically arise from the upper licles have been most extensively studied. The properties of stem cells,
portion of a hair follicle. Interfollicular eccrine glands, on the other TA cells and terminally differentiated cells are listed in Table 2.2.
hand, originate independently. The glandular cords of cells elongate
during the ensuing weeks, and by 7 months EGA the clear cells and Epidermal Stem Cells
mucin-secreting dark cells characteristic of apocrine glands can be visu- The human epidermis renews itself every 4056 days21. This constant
alized. Apocrine glands function transiently during the third trimester turnover is mediated by epidermal stem cells, which reside in the
and subsequently become quiescent in the neonate. Eccrine glands, basal layer together with TA cells22; the latter constitute the majority
however, do not function in utero but mature and begin functioning of basal keratinocytes. Ultimately, TA cells withdraw from the cell
postnatally. cycle and move suprabasally, which initiates the keratinocyte terminal
differentiation program. In non-acral human skin, epidermal stem
Clinical Relevance cells are scattered within the basal layer, where they give rise to epi-
Ectodermal dysplasias are a large, heterogeneous group of genetic dermal proliferative units (EPUs), columns of cells composed of one
disorders that are characterized by developmental abnormalities in two stem cell and its progeny (Fig. 2.4)23. The location of epidermal stem
or more major ectodermal appendages (hair, teeth, nails, and sweat cells in human palmoplantar skin is less clear, but there is some evi-
60 glands); other ectodermal structures (e.g. sebaceous glands) can also be dence that they are found at the base of the rete ridges, where they
affected15 (see Ch. 63). Hypohidrotic ectodermal dysplasia (HED) is a are physically protected from environmental stress. However, other
PROPERTIES OF STEM CELLS, TRANSIT AMPLIFYING CELLS AND TERMINALLY DIFFERENTIATED CELLS

Stem cells Transit amplifying cells Terminally differentiated cells


CHAPTER
Self-renewal* Unlimited Limited No
Potential for differentiation* Multipotent

Limited None 2
Cycling in normal epidermis Slow Rapid None

Skin Development and Maintenance


Proliferative potential (e.g. during fetal development and wound healing) High Limited None

Growth in culture Sustained clonal growth Small abortive clones None
Maintenance of tissue homeostasis Yes Limited No
*In order to maintain both the stem cell and differentiating cell compartments, upon division a stem cell gives rise to another stem cell and a cell that is destined to terminally differentiate
(transit amplifying cell); this is referred to as asymmetry of fate.

Retention of a DNA label (e.g. 5-bromo-2-deoxyuridine or tritiated thymidine) is utilized as an in vivo marker of stem cells based on their slowly cycling nature.

The proliferative potential of transit amplifying cells of the matrix is higher than those of the interfollicular epidermis, but still finite; the former cells can be serially passaged in culture.

Table 2.2 Properties of stem cells, transit amplifying cells and terminally differentiated cells.

EPIDERMAL STEM CELLS

A B C

Cornified
layer

Granular
layer

Spinous
layer

Basal
layer

Differentiating cells

Transit amplifying cells Stem cell

Fig. 2.4 Epidermal stem cells. A Epidermal stem cells are located in the basal layer of the epidermis. Each epidermal stem cell and its progeny forms a vertical
column of progressively differentiating cells, which is known as the epidermal proliferation units (EPU) (blue). B EPUs in human skin grafted onto the back of a
nude mouse. A subset of epidermal keratinocytes was transduced with a -galactosidase-expressing virus. Forty weeks after grafting these cells onto nude mice,
EPUs were visible. Since only epidermal stem cells and not TA cells or differentiated cells are able to persist for this length of time, the presence of EPUs indicates
that they are derived from and maintained by epidermal stem cells. C With each cell division, another stem cell and a transit amplifying (TA) cell are generated
(asymmetry of fate). The TA cells undergo a few rounds of cell division before they permanently withdraw from the cell cycle and initiate the terminal
differentiation program. Reprinted with permission from Kolodka TM, Garlick JA, Taichman LB (1998), Evidence for keratinocyte stem cells in vitro: Long term engraftment and persistence of transgene expression
from retrovirus-transduced keratinocytes Proceedings of the National Academy of Sciences of the United States of America 95:4356-4361, 1998 National Academy of Sciences, U.S.A.

data suggest that palmoplantar epidermal stem cells are located at the potential as well as being self-renewing and multipotent. In vivo, they
top of deep rete ridges. proliferate only when the hair follicle re-enters the active growing phase
One distinguishing feature of stem cells is that they can be main- of anagen, in which the bottom two-thirds of the follicle must be regen-
tained in culture virtually indefinitely. A subset of epidermal basal cells erated. Such appendageal stem cells provide an important ectodermal
forms highly proliferative colonies that can be passaged long term reserve, allowing temporary reconstitution of the surface epidermis
in vitro; referred to as holoclones, they presumably represent stem after severe wounds such as second-degree burns.
cells24. Holoclones can reconstitute epidermis in vitro, and they have In addition to bulge cells, other cell populations in the hair follicle
been utilized for the treatment of burns and inherited skin disorders display stem cell characteristics. One of these populations is located in
(see below). The properties of cutaneous stem cells, including their the lower outer root sheath in anagen hair follicles and is marked by
molecular markers, are summarized in Table 2.3. the expression of Lgr526. Whether these cells represent a separate stem
cell population or whether they are derived from bulge stem cells (or
vice versa) remains to be determined.
Hair Follicle Stem Cells
The hair follicle bulge, which is located in the mid portion of the follicle
(just deep to the sebaceous gland and adjacent to the arrector pili Stem Cell Plasticity
muscle attachment site), is thought to harbor hair follicle stem cells Under homeostatic conditions, epidermal and hair follicle stem
(Fig. 2.5). The bulge represents the lowest permanent portion of the cells only contribute to their respective tissues of origin. However,
hair follicle, and it contains morphologically undifferentiated cells25. bulge-derived keratinocytes can also contribute to the repair of the inter- 61
Hair follicle bulge cells are slow-cycling and have a high proliferative follicular epidermis in response to injury25. Within the healing
CUTANEOUS STEM CELLS

Stem cell Location (niche) Tissue regeneration in Tissue regeneration in injury repair Markers
SECTION homeostasis

1 Keratinocyte
Interfollicular Basal layer Interfollicular epidermis Hair follicle Delta 1, MCSP, 6* and 1 integrins, LRIG1; low
Overview of Basic Science

epidermis Sebaceous gland levels of CD71


Interfollicular epidermis
Hair follicle** Bulge Hair follicle Hair follicle Keratin 15, 6 and 1 integrins, CD200,
Sebaceous gland PHLDA1, follistatin, frizzled homolog 1, LGR5;
Interfollicular epidermis low levels of CD24, CD34, CD71, CD146
Sebaceous At or near the bud of the Sebaceous gland Sebaceous gland Blimp1
gland sebaceous gland
Merkel cell Touch domes of hairy skin Interfollicular epidermis Interfollicular epidermis 6 integrin*, SCA1, CD200
Melanocyte Lower bulge and Hair matrix and Hair matrix and epidermal melanocytes Pax3, TYRP2; negative for KIT, tyrosinase, TYRP1
sub-bulge region epidermal melanocytes
Neural crest-like Bulge (form spheres) ? In vitro: melanocytes, neurons, smooth Twist, SLUG, SOX10, BMI-1, Notch1
muscle, adipocytes, chondrocytes
Dermal Dermis (form spheres) ? In vitro: melanocytes, neurons, Schwann cells, NGFRp75, nestin, OCT4, SLUG
smooth muscle adipocytes, chondrocytes
Skin-derived Dermal papilla (form ? In vitro: neurons, Schwann cells, smooth Nestin, OCT4, SLUG, Twist
precursors spheres) muscle adipocytes
*Also expressed by epidermal transit amplifying cells.
**Additional stem cell populations identified within the hair follicle in mice include LGR6-positive cells in the central isthmus and LRIG1-positive cells in the upper isthmus, both of which
contribute to the interfollicular epidermis and sebaceous glands during homeostasis.

Within the outer root sheath just below the sebaceous gland, at or near the site of insertion of the arrector pili muscle.

Upregulated in mouse bulge cells.

Studied primarily in mice; sebaceous gland stem cells have thus far been shown to be unipotent progenitor cells.

Lowest permanent portion of the hair follicle.

Table 2.3 Cutaneous stem cells. CD24, glycoprotein involved in cell adhesion and signaling; CD34, marker of hematopoietic progenitor cells and endothelial cells;
CD71, transferrin receptor; CD146, melanoma cell adhesion molecule/MUC18/S-Endo-1 antigen; CD200, transmembrane glycoprotein that delivers a negative
immunoregulatory signal (may be involved in maintaining immune tolerance); LGR5/6, leucine-rich repeat-containing G protein-coupled receptor 5/6; LRIG1,
leucine-rich repeats and immunoglobulin-like domains 1 (epidermal growth factor receptor antagonist and regulator of stem cell quiescence); MCSP, melanoma-
associated chondroitin sulfate proteoglycan; NGFR, nerve growth factor receptor; PHLDA1, pleckstrin homology-like domain, family A, member1; SCA1, stem cell
antigen 1; TYRP, tyrosinase-related protein.

KERATINOCYTE AND MELANOCYTE STEM CELLS


epidermis, bulge-derived cells survive only transiently, as they are rapidly
replaced by progenies of epidermal stem cells. Similarly, epidermal stem
Hair shaft
cells can contribute to hair follicle formation in the center of large
wounds27.
Furthermore, emerging data indicate that cells in adult mammals
Epidermis
are far more flexible in their potential to form other cell types than
was previously thought. Some cell types, particularly bone marrow
cells, are able to incorporate themselves into diverse tissues (such as
the epidermis or heart) and to adopt the fates of resident cells, contrib-
Dermis
uting progeny to the tissue. Induced pluripotent stem cells (iPS) are
somatic cells (e.g. fibroblasts) that have been reprogrammed into an
embryonic stem cell-like state28; these cells can be stimulated to dif-
ferentiate into a variety of cell types, including keratinocytes. Thus,
Bulge
many adult cells, including different skin cell types, may be capable of
dedifferentiating into pluripotent stem cells.

Stem Cell-Based Therapy for Genetic Skin Disease


Stem cells have been utilized in several strategies for gene therapy of
heritable skin diseases (Fig. 2.6). Treatment via delivery of gene-
corrected stem cells has the advantage of continuous protein synthesis
in the skin, but it may carry a risk of oncogenesis (especially with
retroviral vectors). In a pilot study, laminin-3-deficient skin from a
man with junctional EB was corrected by transplantation of stem cell-
enriched epidermal grafts transduced ex vivo with a retroviral vector
Epidermal stem cells
encoding normal LAMB3 cDNA. The treatment resulted in expression
of functional laminin 332 and an absence of blistering in the corrected
Hair follicle stem cells
epidermis, both sustained over several years of follow-up. Clinical trials
Melanocyte stem cells
(in lowest permanent are also ongoing to investigate the value of bone marrow-derived stem
portion of hair follicle) cell transplantation as a systemic treatment for the extremely severe
recessive dystrophic form of EB (RDEB). In an initial report29, bone
marrow transplantation after total or partial myeloablation resulted in
62 Fig. 2.5 Keratinocyte and melanocyte stem cells. Note that the bulge is substantial proportions of donor cells in the skin, increased collagen
continuous with the outer root sheath. VII deposition at the dermalepidermal junction and variably decreased
Fig. 2.6 Strategies for stem cell therapy of genetic
STRATEGIES FOR STEM CELL THERAPY OF GENETIC SKIN DISORDERS skin disorders. A One strategy is to isolate epidermal
stem cells from a patient, correct the genetic defect
in vitro (e.g. using a viral vector), and graft epithelial
B Induced pluripotent stem cells sheets containing the corrected stem cells back onto CHAPTER

Patient somatic cells


(e.g. fibroblasts)
the patient. B Gene therapy could also potentially
utilize induced pluripotent stem (iPS) cells, which are
generated from somatic cells (e.g. fibroblasts). The
2

Skin Development and Maintenance


Reprogram somatic genetic defect could be corrected via homologous
A Epidermal stem cells cells into iPS cells recombination in these cells, which would then be
differentiated into keratinocytes and grafted onto the
patient. C A third strategy involves the use of
hematopoietic stem cells (HSC). Upon systemic
Patient epidermal administration (following conditioning), allogeneic
stem cells Patient iPS cells HSC cells have the capacity to home to the skin (e.g.
to sites of injury in patients with epidermolysis
Transduce stem cells with bullosa) and produce differentiated progeny cells
Correct genetic mutation
virus encoding therapeutic
by homologous
that provide the needed skin protein (e.g. collagen
gene ( )
recombination ( ) VII). D Finally, therapeutic genes could be directly
administered to patients via a virus or another vector.
To achieve permanent correction, the vector must
target stem cells that could supply the protein to the
Corrected epidermal skin.
stem cells
Corrected iPS cells
Expand and graft back
onto patient Differentiate corrected iPS
cells into keratinocytes

C Hematopoietic stem cells Corrected keratinocytes

Obtain hematopoietic stem


cells from matched donor

Expand and graft back


onto patient
Hematopoietic
stem cells D Direct delivery

Expand and deliver Virus or other vector


systemically to patient encoding a therapeutic
following conditioning gene; for long-lasting effects,
could target stem cells that
would supply the skin with
the required protein

blistering in children with RDEB. Other investigators are studying for the known family mutation(s); unaffected embryos are then selected
alternative sources of stem cells for the treatment of EB. for uterine implantation. For X-linked disorders, sex determination has
been utilized both in conjunction with specific genetic analysis and to
identify embryos of a particular sex for selective transfer. Preimplanta-
PRENATAL DIAGNOSIS OF GENODERMATOSES tion genetic diagnosis obviates the need to terminate a pregnancy with
an affected fetus. However, it has several disadvantages compared to
chorionic villus sampling or amniocentesis, including higher cost,
Many genodermatoses are incompatible with survival to term or are
technical difficulties (e.g. contamination by extraneous DNA), and a
associated with significant morbidity or even mortality after birth,
low rate of completed pregnancies.
making prenatal diagnosis desirable. In the early 1980s, fetal skin
biopsy became the first technique available for prenatal diagnosis of
inherited skin diseases. Fetal biopsies are typically obtained between
19 and 22 weeks EGA using ultrasound guidance and then analyzed SIGNIFICANCE OF SKIN DEVELOPMENT
via light and electron microscopy for the characteristic morphologic IN POSTNATAL LIFE
changes associated with the disease in question. The Herlitz form of
junctional EB and epidermolytic hyperkeratosis were the first diseases Understanding skin development is essential for the appropriate diag-
diagnosed prenatally using this technique. nosis and management of congenital skin disorders. This knowledge is
As the causative genes for many genodermatoses have been discov- useful far beyond the neonatal period, however, as many of the regula-
ered, DNA-based testing using material obtained from chorionic villus tory pathways central to development are recapitulated in postnatal life.
sampling (1012 weeks after the last menstrual period/810 weeks For example, PTCH is essential for postnatal hair cycling as well as
EGA) or amniocentesis (1416 weeks after the last menstrual period/12 hair follicle development. In addition to the germline PTCH mutations
14 weeks EGA) has largely replaced fetal skin biopsy, as these tech- that result in the nevoid basal cell carcinoma syndrome, somatic PTCH
niques can be performed earlier and pose less risk to the mother and mutations represent a pathogenic factor in sporadic basal cell carcino-
fetus. The pathogenic mutation(s) must be identified in family members mas. Elucidation of the mechanisms controlling angiogenesis during
prior to prenatal testing. intrauterine development holds promise for improving wound healing
Preimplantation genetic diagnosis is a recently introduced technique and developing new anticancer therapies. Furthermore, delineating the
that allows for prenatal diagnosis before the embryo is implanted and factors involved in wound healing in embryos may point to interven-
pregnancy begins. This technique requires the use of in vitro fertiliza- tions that may decrease scarring in children and adults. Thus, advanc-
tion. One or two cells are taken from the embryo at the blastocyst (6- to ing the understanding of skin embryology may lead to life-saving or 63
10-cell) stage. The cellular DNA is amplified using PCR and analyzed life-enhancing therapies.
REFERENCES
1. Koster MI, Roop DR. Mechanisms regulating epithelial 9. Smith LT, Holbrook KA. Development of dermal 19. Shimomura Y, Agalliu D, Vonica A, et al. APCDD1 is a
stratification. Annu Rev Cell Dev Biol. 2007;23:93113. connective tissue in human embryonic and fetal skin. novel Wnt inhibitor mutated in hereditary hypotrichosis
SECTION
2. Holbrook KA. Structure and function of the developing Scan Electron Microsc. 1982;(Pt 4):174551. simplex. Nature. 2010;464:10437.

1 human skin. In: Goldsmith LA (ed). Physiology,


Biochemistry, and Molecular Biology of the Skin, New
10. Wang X, Reid Sutton V, Omar Peraza-Llanes J, et al.
Mutations in X-linked PORCN, a putative regulator of
20. High A, Zedan W. Basal cell nevus syndrome. Curr Opin
Oncol. 2005;17:1606.
York: Oxford University Press, 1991:63110. Wnt signaling, cause focal dermal hypoplasia. Nat 21. Halprin KM. Epidermal turnover time a
Overview of Basic Science

2a. Holbrook KA, Odland GF. Structure of the human fetal Genet. 2007;39:8368. re-examination. Br J Dermatol. 1972;86:149.
hair canal and initial hair eruption. J Invest Dermatol. 11. Fine JD, Smith LT, Holbrook KA, et al. The appearance 22. Kaur P. Interfollicular epidermal stem cells: identification,
1978;71:38590. of four basement membrane zone antigens in challenges, potential. J Invest Dermatol.
3. Akiyama M, Shimizu H. An update on molecular aspects developing human fetal skin. J Invest Dermatol. 2006;126:14508.
of the non-syndromic ichthyoses. Exp Dermatol. 1984;83:669. 23. Strachan LR, Ghadially R. Tiers of clonal organization in
2008;17:37382. 12. Uitto J, Richard G. Progress in epidermolysis bullosa: the epidermis: the epidermal proliferation unit revisited.
4. Kalia YN, Nonato LB, Lund CH, et al. Development of genetic classification and clinical implications. Am J Med Stem Cell Rev. 2008;4:14957.
skin barrier function in premature infants. J Invest Genet C Semin Med Genet. 2004;131C:6174. 24. Barrandon Y, Green H. Three clonal types of keratinocyte
Dermatol. 1998;111:3206. 13. Mikkola ML. Genetic basis of skin appendage with different capacities for multiplication. Proc Natl
5. Adameyko I, Lallemend F, Aquino JB, et al. Schwann cell development. Semin Cell Dev Biol. 2007;18:22536. Acad Sci U S A. 1987;84:23026.
precursors from nerve innervation are a cellular origin of 14. Millar SE. Molecular mechanisms regulating hair follicle 25. Cotsarelis G. Epithelial stem cells: a folliculocentric view.
melanocytes in skin. Cell. 2009;139:36679. development. J Invest Dermatol. 2002;118:21625. J Invest Dermatol. 2006;126:145968.
6. Schuster C, Vaculik C, Fiala C, et al. HLA-DR+ leukocytes 15. Priolo M, Lagana C. Ectodermal dysplasias: a new 26. Haegebarth A, Clevers H. Wnt signaling, Lgr5, and
acquire CD1 antigens in embryonic and fetal human clinical-genetic classification. J Med Genet. stem cells in the intestine and skin. Am J Pathol.
skin and contain functional antigen-presenting cells. J 2001;38:57985. 2009;174:71521.
Exp Med. 2009;206:16981. 16. Casal ML, Lewis JR, Mauldin EA, et al. Significant 27. Ito M, Yang Z, Andl T, et al. Wnt-dependent de novo
7. Van Keymeulen A, Mascre G, Youseff KK, et al. Epidermal correction of disease after postnatal administration hair follicle regeneration in adult mouse skin after
progenitors give rise to Merkel cells during embryonic of recombinant ectodysplasin A in canine X-linked wounding. Nature. 2007;447:31620.
development and adult homeostasis. J Cell Biol. ectodermal dysplasia. Am J Hum Genet. 2007;81:10506. 28. Zhao R, Daley GQ. From fibroblasts to iPS cells: induced
2009;187:91100. 17. Gaide O, Schneider P. Permanent correction of an pluripotency by defined factors. J Cell Biochem.
8. Dessinioti C, Stratigos AJ, Rigopoulos D, et al. A review inherited ectodermal dysplasia with recombinant EDA. 2008;105:94955.
of genetic disorders of hypopigmentation: lessons Nat Med. 2003;9:61418. 29. Wagner JE, Ishida-Yamamoto A, McGrath JA, et al. Bone
learned from the biology of melanocytes. Exp Dermatol. 18. Koster MI, Roop DR. p63 and epithelial appendage marrow transplantation for recessive dystrophic
2009;18:7419. development. Differentiation. 2004;72:36470. epidermolysis bullosa. N Engl J Med. 2010;363:62939.

64

Você também pode gostar