Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Harper's Textbook of Pediatric Dermatology
Harper's Textbook of Pediatric Dermatology
Harper's Textbook of Pediatric Dermatology
Ebook10,189 pages93 hours

Harper's Textbook of Pediatric Dermatology

Rating: 0 out of 5 stars

()

Read preview

About this ebook

The third edition of this highly regarded text continues to provide a comprehensive resource for pediatric dermatologists. The book offers evidence-based diagnosis and treatment recommendations and covers both common and rare conditions, including emerging conditions and research, especially at the genetic level. A refreshing new text design makes the book more accessible, and new editors and contributors bring a distinctly international perspective to the work.
LanguageEnglish
PublisherWiley
Release dateJul 28, 2011
ISBN9781444345377
Harper's Textbook of Pediatric Dermatology

Related to Harper's Textbook of Pediatric Dermatology

Related ebooks

Medical For You

View More

Related articles

Reviews for Harper's Textbook of Pediatric Dermatology

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Harper's Textbook of Pediatric Dermatology - Prof. Dr. Alan D. Irvine

    Acknowledgements

    The editors wish to acknowledge the work of the three founding editors, John Harper, Arnold Oranje and Neil Prose, who expertly developed the first two editions of this book and established it as an important reference work in paediatric dermatology. We would like to thank the patients and their families who gave permission for their photographs to be reprinted; these images greatly enhance the book. Our patients provide the motivation for this work. It is our great hope that the knowledge presented here will improve patient care in paediatric dermatology around the world. We also wish to thank the many people involved in the production team at Wiley-Blackwell.

    ADI

    PH

    AY

    I was very fortunate to have wonderful mentors early in my career in paediatric dermatology. Through their excellence, these physicians inspired me to develop clinical and research interests in childhood skin disease: Ann Bingham, John Harper, David Atherton, Amy Paller, Tony Mancini and Annette Wagner.

    ADI

    John Harper is not only one of the founders of this textbook, but one of the godfathers of paediatric dermatology as a speciality. I am very grateful for his encouragement, inspiration and support.

    PH

    I am grateful to exceptional mentors and wonderful teachers from whom I have learned much throughout my training and career. Their examples continue to inspire me to work harder to be a better physician. Among them, my thanks go especially to my parents who were my first teachers, as well as Paul Honig, Lawrence Eichenfield, William James, Julie Francis and the late Walter Tunnessen.

    AY

    List of Abbreviations

    AA

    alopecia areata

    AAD

    American Academy of Dermatology

    ABC

    ATP-binding cassette

    ACA

    anticentromere antibodies

    ACC

    aplasia cutis congenita

    ACE

    angiotensin-converting enzyme

    ACR

    American College of Rheumatology

    ACTH

    adrenocorticotropic hormone

    AD

    atopic dermatitis

    AD

    autosomal dominant

    ADA

    adenosine deaminase

    ADHD

    attention deficit hyperactivity disorder

    ADR

    adverse drug reaction

    AEDS

    atopic eczema/dermatitis syndrome

    AGA

    androgenetic alopecia

    AGEP

    acute generalized exanthematous pustulosis

    aGVHD

    acute GVHD

    AGM

    acrylate gelling material

    AHA

    antihistone antibody

    AHO

    Albright’s hereditary osteodystrophy

    AIDS

    acquired immune deficiency syndrome

    ALL

    acute lymphoblastic leukaemia

    ALT

    alanine transaminase

    AML

    acute myeloid leukaemia

    AMMoL

    acute myelomonocytic leukaemia

    AMoL

    acute monocytic leukaemia

    ANA

    antinuclear antibody

    AP

    adaptor protein complex

    APC

    antigen-presenting cell

    APECED

    autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (syndrome)

    APSS

    acral peeling skin syndrome

    AR

    autosomal recessive

    ARCI

    autosomal recessive congenital ichthyosis

    ARD

    adult Refsum disease

    AR-HIES

    autosomal recessive hyperimmunoglobulin E syndrome

    AST

    aspartate transaminase

    AT

    ataxia telangiectasia

    ATG

    anti-thymocyte globulin

    ATGL

    adipose triglyceride lipase

    ATPase

    adenosine triphosphatase

    AUC

    area under the concentration-time curve

    AZA

    azathioprine

    BAL

    bronchoalveolar lavage

    BCC

    basal cell carcinoma

    BCG

    bacille Calmette–Guérin

    BCIE

    bullous congenital ichthyosiform erythroderma

    BDNG

    British Dermatological Nursing Group

    BMP

    bone morphogenetic protein

    BMT

    bone marrow transplants

    BO

    branchio-otic

    BOR

    branchio-oto-renal

    BP

    blood pressure

    CAM

    cell adhesion molecule

    C-ALCL

    cutaneous anaplastic large cell lymphoma

    CBCL

    cutaneous B-cell lymphoma

    CCC

    congenital cutaneous candidiasis

    CDP

    chondrodysplasia punctata

    CDPX2

    X-linked dominant chondrodysplasia punctata

    CE

    cell envelope

    CEA

    carcinoembryonic antigen

    CEDNIK

    cerebral dysgenesis, neuropathy, ichthyosis, keratoderma (syndrome)

    CFC

    cardiofaciocutaneous syndrome

    CGD

    chronic granulomatous disease

    CGRP

    calcitonin gene-related product

    CGS

    contiguous gene syndrome

    CHAND

    curly hair, ankyloblepheron, nail dysplasia (syndrome)

    CHH

    Conradi–Hünermann–Happle syndrome

    CHH

    cartilage–hair hypoplasia

    CHILD

    congenital hemidysplasia with ichthyosiform erythroderma and limb defects (syndrome)

    CHIME

    colobomas, congenital heart disease, early-onset ichthyosiform dermatosis, mental retardation and ear abnormalities

    CHS

    Chédiak–Higashi syndrome

    CIE

    congenital ichthyosiform erythroderma

    CLM

    cutaneous larva migrans

    CLOVE

    congenital lipomatous overgrowth, vascular malformations and epidermal naevi (syndrome)

    CMC

    chronic mucocutaneous candidiasis

    CML

    chronic myeloid leukaemia

    CMN

    congenital mesoblastic nephroma

    CMTC

    cutis marmorata telangiectatica congenita

    CMV

    cytomegalovirus

    CNS

    central nervous system

    CNTP

    connective tissue naevus of the proteoglycan type

    CNV

    chromosome copy number variations

    CRP

    C-reactive protein

    CSF

    cerebrospinal fluid

    CSMH

    congenital smooth muscle hamartoma

    CT

    computed tomography

    CTCL

    cutaneous T-cell lymphoma

    CTGF

    connective tissue growth factor

    CTL

    cytotoxic T-lymphocyte

    CTLA-4

    cytotoxic T-lymphocyte antigen-4

    CVG

    cutis verticis gyrata

    CVI

    common variable immunodeficiency

    Cx

    connexin

    DBPCDC

    double-blind placebo-controlled drug challenge

    DEJ

    dermoepidermal junction

    DFSP

    dermatofibrosarcoma protuberans

    DHEA

    dihydroepiandrosterone

    DIHS/AHS

    drug-induced or anticonvulsant hypersensitivity syndrome

    DMARD

    disease modifying antirheumatic drugs

    DMSO

    dimethyl sulphoxide

    DOC

    disorders of cornification

    DOPA

    dihydroxyphenylalanine

    DRESS

    drug rash with eosinophilia and systemic symptoms

    DSAP

    disseminated superficial actinic porokeratosis

    DSP

    disseminated superficial porokeratosis

    DTE

    desmoplastic trichoepithelioma

    EAACI

    European Academy of Allergy and Clinical Immunology

    EBP

    emopamil binding protein

    EBV

    Epstein–Barr virus

    EBS-DM

    epidermolysis bullosa simplex Dowling–Meara

    ECF

    eosinophil chemotactic factor

    ECP

    extracorporeal photopheresis

    EDP

    erythema dyschromicum perstans

    EEG

    electroencephalogram

    EGA

    estimated gestational age

    EGF

    epidermal growth factor

    EHK

    epidermolytic hyperkeratosis

    EKA

    erythrokeratoderma with ataxia

    EKC

    erythrokeratoderma en cocardes

    EKV

    erythrokeratoderma variabilis

    ELISA

    enzyme-linked immunosorbent assay

    EMLA

    eutectic mixture of local anaesthetics

    EN

    epidermal naevus

    EN-D

    epidermal naevus – Darier type

    ENDA

    European Network for Drug Allergy

    EOS

    early-onset sarcoidosis

    EP

    eccrine poroma

    ERA

    enthesitis-related arthritus

    ESPD

    European Society of Pediatric Derrmatology

    ESR

    erythrocyte sedimentation rate

    ETN

    erythema toxicum neonatorum

    EULAR

    European League Against Rheumatism

    EV-HPV

    epidermodysplasia verruciformis-associated human papillomavirus

    FADH

    fatty alcohol dehydrogenase

    FALDH

    fatty aldehyde dehydrogenase

    FATP

    fatty acid transport protein

    FDA

    Food and Drug Administration (USA)

    FDE

    fixed drug eruptions

    FFD

    Fox-Fordyce disease

    FFM

    focus-floating microscopy

    FGFR

    fibroblast growth factor receptor

    FISH

    fluorescence in situ hybridization

    FITC

    fluorescein isothiocynate

    FMF

    familial Mediterranean fever

    FOP

    fibrodysplasia ossificans progressive

    FTC

    familial tumoral calcinosis

    FTG

    full-thickness skin graft

    5-FU

    5-fluoracil

    GABA

    γ-aminobutyric acid

    GBFDE

    generalized bullous fixed drug eruption

    GCDFP

    gross cystic disease fluid protein

    GFR

    glomerular filtration rate

    GI

    gastrointestinal

    GM-CSF

    granulocyte-macrophage colony-stimulating factor

    GMS

    Gomori’s methenamine silver

    GNCST

    granular nerve cell sheath tumour

    GOSH

    Great Ormond Street Hospital for Children

    GPP

    generalized pustular psoriasis

    GS

    Griscelli syndrome

    GVH

    graft-versus-host

    GVHD

    graft-versus-host-disease

    GVHR

    graft-versus-host reaction

    GVL

    graft-versus-leukaemia

    HAE

    hereditary angioedema

    HCC

    harlequin colour change

    HCG

    human chorionic gonadotropin

    HCT

    hemopoietic cell transplantation

    HDL

    high-density lipoprotein

    HHD

    Hailey–Hailey disease

    HI

    harlequin ichthyosis

    HID

    hystrix-like ichthyosis with deafness (syndrome)

    HIES

    hyperimmunoglobulin E syndrome

    HIMS

    hyperimmunoglobulin M syndrome

    HIP

    helix initiation peptide

    HIV

    human immunodeficiency virus

    HLA

    human leucocyte antigen

    HPC

    haemangiopericytoma

    HPS

    Hermansky-Pudlak syndrome

    HPETE

    hydroperoxyeicosatetraenoic acid

    HPV

    human papillomavirus

    HS

    hidradenitis suppurativa

    HSV

    herpes simplex virus

    HTLV

    human T-lymphotropic type 1

    HTP

    helix termination peptide

    IBIDS

    ichthyosis, brittle hair, intellectual impairment, decreased fertility and short stature

    IBS

    ichthyosis bullosa of Siemens

    ICAM

    intracellular adhesion molecule

    ICD

    irritant contact dermatitis

    IF

    infantile fibrosarcoma

    IFAP

    ichthyosis follicularis with atrichia and photophobia

    IFN

    interferon

    Ig

    immunoglobulin

    IgA1

    IgA subtype 1

    IgM

    immunoglobulin M

    IGF

    insulin-like growth factor

    IGFBP

    IGF binding protein

    IGRA

    interferon-gamma release assay

    IgεRI

    high-affinity IgE receptor

    IHCM

    ichthyosis hystrix of Curth–Macklin

    HIS

    ichthyosis hypotrichosis syndrome

    IHSC

    ichthyosis-hypotrichosis-sclerosing cholangitis (syndrome)

    IL

    interleukin

    IL-1β

    interleukin-1β

    ILAR

    International League of Associations for Rheumatology

    ILC

    ichthyosis linearis circumflexia

    ILVEN

    inflammatory linear verrucous epidermal naevus

    IPEX

    immune dysregulation, polyendocrinopathy, enteropathy, X-linked

    IPS

    ichthyosis prematurity syndrome

    ISAAC

    International Study of Asthma and Allergies in Childhood

    ISD

    infantile seborrhoeic dermatitis

    IV

    ichthyosis vulgaris

    IVIG

    intravenous immunoglobulin

    JDM

    juvenile dermatomyositis

    JIA

    juvenile idiopathic arthritis

    JSPD

    Japanese Society for Pediatric Dermatology

    KFSD

    keratosis follicularis spinulosa decalvans

    KID

    keratitis, ichthyosis and deafness (syndrome)

    KIF

    keratin intermediate filaments

    KLK

    kallikrein

    KTS

    Klippel–Trenaunay syndrome

    KWE

    keratolytic winter erythema

    LAD

    leucocyte adhesion deficiency

    LAS

    loose anagen syndrome

    LCD

    liquor carbonis detergens

    LCH

    Langherhans’ cell histiocytosis

    LD

    lymphoedema-distichiasis

    LDF

    laser Doppler flowmetry

    LDL

    low-density lipoprotein

    LEC

    lymphatic endothelial cells

    LEKTI

    lymphoepithelial Kazal-type inhibitor

    LFA-3

    lymphocyte function-associated antigen-3

    LI/CIE

    lamellar ichthyosis/congenital ichthyosiform erythroderma

    LMP1

    latent membrane protein 1

    LMX

    liposomal lignocaine

    LOH

    loss of heterozygosity

    LOSSI

    localized scleroderma severity index

    LOX

    lipoxygenase

    LT-β

    lymphotoxin- β

    LTT

    lymphocyte transformation test

    LyP

    lymphomatoid papulosis

    MAC

    membrane attack complex

    MACS

    magnetic-activated cell sorting

    MAIC

    M. avium-intracellulare complex

    MBL

    mannose-binding lectin

    MBTPS2

    membrane-bound transcription factor protease, site 2

    MC

    mast cells

    MC

    molluscum contagiosum

    MC&S

    microscopy, culture and (antibiotic) sensitivity

    MDM

    minor determinant mixture

    MEDNIK

    mental retardation, enteropathy, deafness, peripheral neuropathy, ichthyosis, keratodermia

    MEDOC

    mendelian disorders of cornification

    mHA

    minor histocompatibility antigen

    MHC

    major histocompatibility complex

    MMF

    mycophenolate mofetil

    MMP

    matrix metalloproteinases

    MPE

    maculopapular exanthems

    MRI

    magnetic resonance imaging

    MRSA

    methicillin-resistant Staphylococcus aureus

    MSD

    multiple sulphatase deficiency

    MSH

    melanocyte-stimulating hormone

    NADPH

    nicotinamide adenine dinucleotide phosphate

    NBD

    nucleotide-binding domain

    NBS

    Nijmegan breakage syndrome

    NBT

    nitroblue tetrazolium

    NB-UVB

    narrow-band UVB

    NCAM

    neural cell adhesion molecule

    ND

    naevus depigmentosus

    NF1

    neurofibromatosis type 1

    NFκB

    nuclear factor κB

    NGCO

    non-gestational ovarian choriocarcinoma

    NGFR

    nerve growth factor receptor

    NICE

    National Institute of Health and Clinical Excellence (UK)

    NICU

    neonatal intensive care unit

    NIH

    National Institutes of Health (USA)

    NISCH

    neonatal ichthyosis-sclerosing cholangitis (syndrome)

    NK

    natural killer (cell)

    NLSD

    neutral lipid storage disease

    N2O

    nitrous oxide

    NPSA

    National Patient Safety Agency (UK)

    NPY

    neuropeptide Y

    NS

    naevus sebaceous

    NS

    Netherton syndrome

    NSAIDs

    non-steroidal anti-inflammatory drugs

    NSV

    non-segmental vitiligo

    NTM

    non-tuberculous mycobacteria

    OA

    ocular albinism

    OC

    osteoma cutis

    OL-EDA-ID

    osteopetrosis, lymphoedema, ectodermal dysplasia anhydrotic and immune deficiency

    O/W

    oil in water

    P1cp

    procollagen type 1 carboxy-terminal peptide

    PA

    phytanic acid

    PAHX

    phytanoyl CoA hydroxylase

    p-ANCA

    perinuclear antineutrophilic cytoplasmic antibody

    PAR2

    protease-activated receptor 2

    PAS

    periodic acid–Schiff

    PASI

    psoriasis area and severity index

    PCFCL

    primary cutaneous follicle-centre lymphoma

    PCFH

    precalcaneal congenital fibrolipomatous hamartoma

    PCLBCL

    primary cutaneous diffuse large B-cell lymphoma

    PCMZL

    primary cutaneous marginal zone B-cell lymphoma

    PCOS

    polycystic ovarian syndrome

    PCR

    polymerase chain reaction

    PCT

    porphyria cutanea tarda

    PCT

    primary care trust

    PDGF

    platelet-derived growth factor

    PDL

    pulsed-dye laser

    PEPD

    paroxysmal extreme pain disorder

    PEN

    porokeratotic eccrine naevus

    PEODDN

    porokeratotic eccrine ostial and dermal duct naevus

    PGP 9.5

    protein gene product 9.5

    PH

    palmoplantar hidradenitis

    PHA

    phytohaemagglutinin

    PHP

    pseudo-hypoparathyroidism

    PhyH

    phytanoyl-CoA 2-hydroxylase

    pI

    isoelectric point

    PI3

    proteinase inhibitor 3

    PIP

    proximal interphalangeal

    PL

    pityriasis lichenoides

    PLC

    pityriasis lichenoides chronica

    PLEVA

    pityriasis lichenoides et varioliformis acuta

    PM

    porokeratosis of Mibelli

    PML

    progressive multifocal leucoencephalopathy

    POEMS

    polyneuropathy, organomegaly, endocrinopathy, M protein, skin changes (syndrome)

    PPAR

    peroxisome proliferator-activated receptor

    PPK

    palmoplantar keratodermas

    PPL

    penicilloyl-polylysine

    POH

    progressive osseous heteroplasia

    PPPD

    porokeratosis palmaris et plantaris disseminata

    PRES

    Paediatric Rheumatology European Society

    PRINTO

    Paediatric Rheumatology International Trials Organization

    PRP

    pityriasis rubra pilaris

    PSEK

    progressive symmetric erythrokeratoderma

    PSH

    premature sebaceous hyperplasia

    PSS

    peeling skin syndromes

    PTC

    premature termination codon

    PTH

    parathyroid hormone

    PTHrP

    parathyroid-hormone-related peptide

    PUVA

    psoralens plus UVA

    PXE

    pseudo-xanthoma elasticum

    PWS

    port wine stain

    QUADAS

    Quality Assessment of Diagnostic Accuracy tool

    RAMBA

    retinioic acid metabolism blocking agent

    RCDP

    rhizomelic chondrodysplasia punctata

    RCT

    randomized controlled trial

    RF

    rheumatoid factor

    RMH

    rhabdomyomatous mesenchymal hamartoma

    ROS

    reactive oxygen species

    RTX

    rituximab

    RXLI

    recessive X-linked ichthyosis

    SCC

    squamous cell carcinoma

    SCCE

    stratum corneum chymotryptic enzyme

    SCID

    severe combined immunodeficiency

    SCT

    stem cell transplantation

    SCTE

    stratum corneum tryptic enzyme

    SD

    seborrhoeic dermatitis

    SEI

    superficial epidermolytic ichthyosis

    SFT

    solitary fibrous tumour

    SHP

    Schönlein–Henoch purpura

    SIB

    self-injurious behaviour

    SID

    sudden infant death (syndrome)

    sIgE

    drug-specific IgE antibodies

    SJS

    Stevens–Johnson syndrome

    SLADP

    Latin American Society for Pediatric Dermatology

    SLC27

    solute carrier family 27

    SLE

    systemic lupus erythematosus

    SLN

    speckled lentiginous naevus

    SLOS

    Smith–Lemli–Opitz syndrome

    SLPI

    secretory leucocyte protease inhibitor

    SLS

    Sjögren–Larsson syndrome

    SNP

    single nucleotide polymorphism

    SP

    syringocystadenoma papilliferum

    SPD

    Society for Pediatric Dermatology

    SPECT

    single-photon emission computerized tomography

    SPF

    sun protection factor

    SPINK

    serine protease inhibitor Kazal type

    SPRR

    small proline-rich proteins

    SPTL

    subcutaneous panniculitis-like T-cell lymphoma

    SSG

    split-thickness skin graft

    SSLR

    serum sickness-like reactions

    SSRI

    selective serotonin reuptake inhibitors

    SSSS

    staphylococcal scalded skin syndrome

    STI

    sexually transmitted infection

    STS

    steroid sulphatase

    TAC

    tetracaine/adrenaline/cocaine

    TBSA

    total body surface area

    TCS

    topical corticosteroids

    TCR

    T-cell receptor

    TEN

    toxic epidermal necrolysis

    TEWL

    transepidermal water loss

    TG

    transglutaminase

    TG

    triacylglycerol

    TG1

    transglutaminase 1

    TGF

    transforming growth factor

    TJ

    tight junction

    TLR

    toll-like receptor

    TMD

    transmembrane domains

    TMP-SMX

    trimethoprim-sulfamethoxazole

    TNF

    tumour necrosis factor

    TPM

    transient pustular melanosis

    TPMT

    thiopurine methyltransferase

    TRAPS

    TNF receptor superfamily 1A-associated periodic fever syndrome

    TRT

    thermal relaxation time

    TS

    tuberous sclerosis

    TTD

    trichothiodystrophy

    UD

    unrelated donor

    uE3

    unconjugated oestriol

    UV

    ultraviolet

    UVB

    ultraviolet light

    VEGF

    vascular endothelial growth factor

    VEGFR3

    vascular endothelial growth factor receptor 3

    VLCFA

    very-long-chain fatty acid

    VZIG

    varicella zoster immunoglobulin

    VZV

    varicella zoster virus

    WAO

    World Allergy Organization

    WAS

    Wiskott-Aldrich syndrome

    WHO

    World Health Organization

    W/O

    water in oil

    XD

    X-linked dominant

    XLMR

    X-linked mental retardation

    XR

    X-linked recessive

    ZIG

    zoster immune globulin

    ZNS

    Zunich neuroectodermal syndrome

    CHAPTER 1

    The History of Paediatric Dermatology

    John Harper

    Paediatric Dermatology, Great Ormond Street Hospital for Children NHS Trust, London, UK

    Ancient origins of paediatric dermatology

    The global establishment of paediatric dermatology as a recognized subspecialty

    Paediatric dermatology in North America (USA and Canada)

    Paediatric dermatology in Latin America

    Paediatric dermatology in Japan

    Paediatric dermatology in Europe

    The future for paediatric dermatology

    Ancient Origins of Paediatric Dermatology

    Visible skin abnormalities have been recognized since the dawn of history, dating back about 5000 years to the oldest medical text of Sumer, an ancient city of Mesopotamia. The text, in the form of a clay tablet, is a pharmacopoeia in which many salves (a medical ointment used to soothe the head or other body surface) and lotions are listed. There are other Mesopotamian medical tablets that mention conditions relating to itching, leprosy, impetigo, erysipelas and jaundice. Specifically in the newborn are recorded the vernix caseosa and some congenital skin abnormalities. Other evidence of early records of skin ailments and their treatment is found in the Egyptian papyri of 1500 BC, in the sacred books of the Hindus of ancient India and in the Hippocratic writings of the Greeks. Hippocrates drew attention to cutaneous disorders in children, restricted largely to clinical observation. In the Hippocratic writings at least six passages deal specifically with skin ailments in children. He refers to leprosy, lichen and leuke. Leuke was considered by many scholars to be a form of leprosy or vitiligo, and lichen has been variously interpreted as representing a variety of skin conditions – ringworm, eczema, psoriasis, herpes – characterized by eruptions and itching.

    Rhazes (865–925), a Persian physician, alchemist, philosopher and scholar, is considered to be the father of paediatrics for writing The Diseases of Children, the first book of paediatrics. He describes infantile eczema (cradle cap), which he called ‘sahafati’ … lesions exuding fluid spread over the head and face causing the child to cry and scratch … he concluded the affliction proceeded from superfluidities of the blood and excess moisture of the skin … he recommended depilation of the scalp followed by the application of atriplex leaves to draw out the ‘poison’ … he also mentioned the use of lead ointment. His classic work was to distinguish smallpox and measles, as well as chickenpox, through his clinical characterization of these diseases. Avicenna (980–1037), whose Canon of Medicine exerted a great influence on medieval medicine, described an eruptive condition now considered to be scarlet fever. Avenzoar (1113–62), another Arabian philosopher, was the first to be credited with describing the itchy mite of scabies.

    The first monograph on dermatology is Galen’s (129–200 AD) De Tumoribus Praeter Naturam on abnormal swellings. In 1572, Geronimo Mercuriali of Forlì, Italy, completed De morbis cutaneis (‘Diseases of the Skin’), and this is recognized as the first scientific work to be dedicated to dermatology.

    Robert Willan (1757–1812) is considered the founder of dermatology as a medical specialty. Thomas Bateman (1778–1821) was a British physician and a pioneer in the field of dermatology. Willan died leaving Bateman to continue and expand on the work of his mentor. In 1817 Bateman published an atlas called Delineations of Cutaneous Disease (Fig. 1.1).

    Fig. 1.1 From Delineations of Cutaneous Diseases. Thomas Bateman. Henry G. Bohn, London 1st edn. 1817; 2nd edn 1840: PLATE 1. The STROPHULUS intertinctus; popularly termed the Red Gum, or Gown; a pimply eruption of a vivid red colour, rising sensibly above the level of the skin, and intermixed often with dots and red patches which have no elevation. It is peculiar to very young infants; and often consistent with good health.

    c01f001

    The first treatise exclusively devoted to paediatric dermatology was Cutaneous Diseases Incidental to Childhood by Walter C. Dendy, published in London in 1827 (Fig. 1.2). The author was Surgeon to the Royal Universal Dispensary for Children, later to become the Royal Waterloo Hospital for Children and Women, part of St Thomas’ Hospital.

    Fig. 1.2 A Treatise on the Cutaneous Diseases Incidental to Childhood. Walter C. Dendy, John Churchill, London, 1827. The first textbook of pediatric dermatology.

    Courtesy of the Wellcome Library, London.

    c01f002

    Two decades later Charles West worked at the same hospital, where only ambulatory patients attended. In 1852 Dr Charles West played a significant role in establishing the Hospital for Sick Children, Great Ormond Street, London.

    In the Western world, the first generally accepted paediatric hospital is the Hôpital des Enfants Malades, opened in Paris in June 1802, on the site of a previous orphanage. From its beginning, this famous hospital accepted patients up to the age of 15 years and it continues to this day as the paediatric division of the Necker-Enfants Malades Hospital, created in 1920 by merger with the Necker Hospital, founded in 1778 for adults.

    The Global Establishment of Paediatric Dermatology As A Recognized Subspecialty

    It was not until 1972 that paediatric dermatology was ‘officially born’ at the first International Symposium of Paediatric Dermatology in Mexico City (Fig. 1.3). Distinguished physicians met at the famous San Angelin Restaurant and founded the International Society of Pediatric Dermatology. These pioneers included: Martin Beare (Ireland); Ferdinando Gianotti (Italy); Joan Hodgeman (USA); Coleman Jacobson (USA); Guinter Kahn (USA); Andrew Margileth (USA); Edmund Moynahan (England); Dagoberto Pierini (Argentina); Ramon Ruiz-Maldonado (Mexico); Lawrence Solomon (USA); Eva Torok (Hungary) and Kazuya Yamamoto (Japan). Prior to this children and adolescents with skin maladies were mainly looked after by paediatricians and primary care physicians, with only a few dermatologists and academics interested in the research and management of these children. Following that historic meeting in Mexico City, interest in this discipline of medicine has grown dramatically throughout the world and is now integral to all major dermatological and paediatric meetings. Since then, 10 World Congresses of Paediatric Dermatology have taken place: Chicago (Fig. 1.4), Monte Carlo, Tokyo, Milan, Toronto, Buenos Aires, Paris, Cancun, Rome and Thailand.

    Fig. 1.3 Poster advertising the first International Symposium on Pediatric Dermatology held in Mexico City in 1973.

    c01f003

    Fig. 1.4 Delegates attending the Second International Congress of Pediatric Dermatology in Chicago, including Ruggero Caputo, Nancy Esterly, Sidney Hurwitz, Alvin Jacobs, Coleman Jacobson, Gunter Kahn, Al Lane, Marc Larregue, Arthur Norins, Jim Rasmussen, Ramon Ruiz Maldonado, Jean-Hilaire Saurat, Laurence Schachner, Lawrence Solomon, Loudes Tamayo, Sam Weinberg, William Weston.

    Courtesy of Dr Susan Bayliss.

    c01f004

    There are now journals specifically dedicated to the subject published in the USA, Japan and Europe. The Society of Pediatric Dermatology in the USA was founded in 1975, the Japanese Society for Pediatric Dermatology in 1977 and the European Society for Pediatric Dermatology in 1983. The past two decades have seen a plethora of textbooks on paediatric dermatology, but it is important to highlight two authors, Sidney Hurwitz and William Weston, whose books respectively had a profound influence on the establishment of the specialty and its teaching. There are two up-to-date encyclopaedic textbooks of pediatric dermatology; one edited by Schachner and Hanson and the other edited by Harper, Oranje and Prose (first two editions) and Irvine, Hoeger and Yan (third edition).

    Mexico has led the way in training with a programme for both paediatricians and dermatologists, founded in 1973 by Ramon Ruiz Maldonado and Lourdes Tamayo. More than 100 specialists now working in most Latin American countries have been trained in that programme. In the USA paediatric dermatology became an independent board-certified subspecialty as recently as 2004. Elsewhere in the world training remains ad hoc and includes paediatricians with a special interest in dermatology, dermatologists with a special interest in children, and a select handful who have a full training in both specialties. In some countries the lack of cooperation between the two disciplines can be a stumbling block to the establishment of the specialty.

    Paediatric Dermatology in North America (USA and Canada)

    Henry Harris Perlman was the first physician to be board certified by both the American Academy of Pediatrics and the American Academy of Dermatology (AAD), the first to limit his practice to diseases of the skin in children (1946) and the first in the USA to write a textbook dedicated solely to Pediatric Dermatology (see Moynahan, 1961, p. 954 for a review).

    In the 1960s and 1970s interest in pediatric dermatology was pursued by a group of physicians in the USA, who made a major contribution to the establishment of the subject as a recognized specialty (in alphabetical order): Carroll Burgoon Jr, Philadelphia; Sidney Hurwitz, New Haven, Connecticut; Alvin Jacobs, Palo Alto, California; Gunter Kahn, Miami, Florida; Peter Koblenzer, Morristown, New Jersey; Arthur Norins, Indianapolis, Indiana; James Rasmussen, Buffalo, New York; Lawrence Solomon and Nancy Esterly, Chicago; Samuel Weinberg, New York City; and William Weston, Denver, Colorado.

    At the AAD meeting in Chicago in December 1974, four paediatric dermatologists met for the purpose of starting a Society for Pediatric Dermatology (SPD). They were Sidney Hurwitz, Sam Weinberg, William Weston and Alvin Jacobs. A meeting was organized in Dallas, Texas, in February 1976, hosted by Coleman Jacobson. Thirty-eight interested in membership attended. Sidney Hurwitz was elected President, Sam Weinberg and Alvin Jacobs Vice-Presidents, and William Weston Secretary-Treasurer (Fig. 1.4). The first Scientific Meeting of the Society was hosted by William Weston in Aspen, Colorado, in July 1976.

    Subsequent annual meetings attracted more and more individuals and now the SPD is recognized internationally to be one of the leading groups representing our specialty worldwide. In addition to the summer annual meeting, the one-day meeting prior to the AAD has been very successful and attracts a high attendance.

    At the 10th annual meeting in Cape Cod, the first annual Sidney Hurwitz lectureship was inaugurated. Dr Tomisaku Kawasaki of Tokyo, who first described the disorder that bears his name, gave an update on Kawasaki’s disease.

    The AAD now recognizes paediatric dermatology as a board certified subspecialty, for which there are designated training programmes with defined certification requirements (http://www.abderm.org/subspecialties/qualification.html).

    The Journal of Pediatric Dermatology was first published in 1983 with Lawrence Solomon and Nancy Esterly as editors. It is the main journal worldwide for the subspecialty with a high profile of clinical and research contributions. It is published by Wiley Blackwell and the current editors are Ilona Frieden and Lawrence Eichenfield.

    Paediatric dermatology in the USA has seen a rapid growth of interest, with many new young paediatric dermatologists (too many to list) who are making significant contributions to our understanding and treatment of skin ailments in children and adolescents. Many of these clinicians and research workers have contributed to this textbook.

    In Canada, paediatric dermatology has been led by Bernice Krafchik in Toronto, with Julie Prendiville (Vancouver) and Julie Powell and Danielle Marcoux (Montreal). In 1981, Bernice Krafchik hosted the meeting of the Society of Pediatric Dermatology, with over 150 attendees from Canada and the USA. In 1992, Bernice Krafchik and Jim Rasmussen organized the sixth World Congress of Pediatric Dermatology in Toronto.

    Paediatric Dermatology in Latin America

    Modern day paediatric dermatology owes much to the dedication, enthusiasm and commitment of Ramon Ruiz Maldonado and Lourdes Tamayo of Mexico City in establishing paediatric dermatology as a subspecialty, not just in Mexico but throughout Latin America. Their influence, in promoting and supporting paediatric dermatology, has been worldwide. The first Symposium of Pediatric Dermatology was in 1972 in Mexico City (Fig. 1.3) and at that time the International Society of Pediatric Dermatology was founded. The first training programme in the world was established at the National Institute of Pediatrics in Mexico City in 1973, for both paediatricians and dermatologists. Certified paediatricians do 3 years of training in paediatric dermatology, and certified dermatologists do 1 year’s training in paediatric dermatology to obtain a diploma in paediatic dermatology.

    Subsequently, Dagoberto Pierini launched the Paediatic Dermatology training programme in Argentina, where there are currently two training institutes, headed by Adrian Pierini and Margarita Larralde. The seventh World Congress of Pediatric Dermatology was held in Buenos Aires under the Presidency of Adrian Pierini.

    Paediatic dermatology is now a recognized subspecialty in most Latin American countries. Past and present leading paediatric dermatologists include: Evelyne Halpert (Colombia); Hector Caceres, Leonardo Sanchez (Peru); Luiz Alfredo Gonzales Aveledo (Venezuela); Adrian Pierini, Margarita Larralde, Rita Garcia Diaz, Maria Rosa Cordisco, Jose Antonio Massimo (Argentina); Ramon Ruiz Maldonado, Lourdes Tamayo, Carola Duran McKinster (Mexico); Fausto Forin Alonso, Susana Giraldi (Brazil); Julia Oroz, Winston Martinez, Sergio Silva (Chile); and Raul Vignale, Marcelo Ruvertoni (Uruguay).

    In 1996 in Lima, the Latin American Society for Pediatric Dermatology (SLADP) was founded. The first Congress was held in Bogota in 1997, organized by Evelyne Halpert. The SLADP has grown significantly over the years and now includes representation of 15 countries, meeting every 3 years. Hector Caceres and his working group are responsible for the journal Revista de Dermatología Pediátrica Latinoamericana.

    Paediatric Dermatology in Japan

    Kazuya Yamamoto was Head of the Department of Dermatology at the National Children’s Hospital in Tokyo and in 1977 established the Japanese Society for Pediatric Dermatology (JSPD). The first President was Professor Toshiaki Yasuda. The JSPD met annually and at each meeting invited both national and international guest speakers. The number of members of the JSPD has risen to over 1000. In 1986 Tokyo hosted the fourth World Congress of Pediatric Dermatology, with Professor Harukuni Urabe as President and Kazuya Yamamoto as the Head of the Secretariat. The Journal of the JSPD was first published in 1982 and has continued biannually to the present time.

    Paediatric Dermatology in Europe

    This new medical discipline has become established in most European countries over the past 25 years. Physicians who have made major contributions to paediatric dermatology in Europe include: Jean-Hilaire Saurat, Marc Larregue, Jean Maleville, Yves de Prost and Alain Taieb (France); Ruggero Caputo, Carlo Gelmetti, Giuseppe Fabrizi and Ernesto Bonifazi (Italy); Rudolph Happle, Heiko Traupe (Germany); Martin Beare, Edmund Moynahan, Charles Wells, David Atherton and John Harper (UK); Micheline Song and Linda de Raeve (Belgium); Arnold Oranje, Flora de Waard-van der Spek and Henk Sillevis Smit (The Netherlands); Talia Kakourou (Greece); Juan Ferrando, Ramon Grimalt, Antonio Torrelo (Spain); and Daniel Hohl (Switzerland). This list does not include a number of eminent physicians who predate the past 25 years, physicians in other European countries and new young doctors trained in paediatric dermatology.

    National societies have been established in many European countries: Belgium, Croatia, France, Germany, Greece, Hungary, Italy, The Netherlands, Portugal, Spain, Switzerland, Turkey and the UK.

    Research in Europe has made major contributions to our understanding and treatment of skin conditions in children. Examples include: acrodermatitis enteropathica and zinc deficiency (Edmund Moynahan); graft-versus-host disease of the skin (Jean-Hilaire Saurat); the histiocytoses (Ruggero Caputo) and most recently propranolol treatment for haemangiomas (Alain Taieb).

    The European Society of Pediatric Derrmatology (ESPD) was established in 1983. European Congresses have been held in: Munster, 1984; Bari, 1987; Bordeaux, 1990; Bournemouth, 1993; Rotterdam, 1996; Rome, 1999; Barcelona, 2002; Budapest, 2005; and Athens, 2008.

    The first paediatric dermatology course in Europe was set up in 1977 by Marc Larregue and Jean Maleville, held in Arcachon in France and continued annually. Other courses have since been established in Paris, Bari, Rome, Rotterdam, Dundee and Birmingham.

    As well as the journal Pediatric Dermatology to which the ESPD is affiliated, the first edition of Pediatric Dermatology News, edited by Ernesto Bonifazi, was in 1982. In 1991 this publication became the European Journal of Pediatric Dermatology.

    The Future for Paediatric Dermatology

    The inspired vision of our predecessors has firmly established paediatric dermatology as a recognized subspecialty internationally. Compared to other medical subspecialties, paediatric dermatology is in its infancy in development. The main issue that needs addressing is an agreed international training programme. This has been achieved in Mexico and in the USA and must be a priority for the International Society for Pediatric Dermatology (ISPD) as we strive forward in this new millennium. To quote Sidney Hurwitz, in Pediatric Dermatology in 1988: ‘Those of us who have committed ourselves to this discipline are proud of the accomplishments of the past, appreciate the rapidly growing interest in this field in the present and look forward to the challenges of the future with continued optimism and enthusiasm.’

    Further reading

    Copeman PWM. The creation of global dermatology. J Roy Soc Med 1995;88:78–84.

    Galimberti R, Pierini AM, Cervini AB. (eds) History of Latin American Dermatology. Toulouse: Edicions Privat, 2007.

    Hurwitz S. The history of pediatric dermatology in the United States. Pediatr Dermatol 1988;5:280–5.

    Moynahan EJ. Review of Pediatric Dermatology by Henry H Perlman. BMJ;1961(1 April):954.

    Radbill SX. Pediatric dermatology in antiquity: part I. Int J Dermatol 1975;14:363–8.

    Radbill SX. Pediatric dermatology in antiquity: part II. Roman Empire. Int J Dermatol 1976;15:303–7.

    Radbill SX. Pediatric dermatology in antiquity: part III. Int J Dermatol 1978;17:427–34.

    Radbill SX. Pediatric dermatology: chronological excursions into the literature. Part I. Pediatric dermatology in general medical texts. Int J Dermatol 1985;26:250–6.

    Radbill SX. Pediatric dermatology: chronological excursions into the literature. Part II. Pediatric dermatology in pediatric texts. Int J Dermatol 1987;26:324–31.

    Radbill SX. Pediatric dermatology: chronological excursions into the literature. Part III. DermatologicTexts. Int.J Dermatol. 1987; 26(6):394–400.

    Radbill SX. Pediatric Dermatology: chronological excursions into the literature. Part IV. Pediatric dermatology texts. Int J Dermatol 1987;26:474–9.

    Ruiz-Maldonado R. Pediatric dermatology accomplishments and challenges for the 21st century. Arch Dermatol 2000;136:84.

    Taieb A, Larregue M, Maleville J. The development of paediatric dermatology. In: Wallach D, Tilles G (eds) Dermatology in France. Toulouse: Editions Privat, 2002; 127–31. http:www.bium.univ-paris5.fr/histmed/medica/cote?extwall00001.

    Yamamoto K. The Society: early days. J Jpn Soc Pediatr Dermatol 2009;28:158–162 [in Japanese].

    CHAPTER 2

    Embryogenesis of the Skin

    Karen A. Holbrook

    Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, USA

    Introduction

    Time-scale of skin development

    Embryonic skin

    Embryonic–fetal transition

    Fetal skin

    Unique features of developing human skin

    Conclusion

    Introduction

    The skin is an ideal organ in which to study development through ageing because it is readily accessible for observation, sampling and evaluation. As an interface, it straddles the internal, systemic world of the individual and the external environment and is vulnerable to and can be modified by both. The skin itself is a remarkably complicated and complex organ, with the normal structure and function of each ‘part’ highly dependent upon what happens in other parts of the skin. In other words, one cannot understand, for example, changes that occur in the epidermis without understanding the nature of the dermis since the dermis has major influences on the activities and functions of the epidermis. This is the case for each region or structure of the skin.

    Development, however, offers an opportunity to study skin structure and function under more ‘controlled’ conditions because, except for changes in the composition of the amniotic fluid (similar to maternal plasma in the embryo but more characteristic of fetal urine in the second trimester [1,2]), the environment of the developing skin is reasonably constant (controlled light, temperature, pressure, etc.). It is possible therefore to investigate how the properties of the different regions and structures of the skin are coordinately established, presumably under the directions of a genetic program, and spared from the assaults of the external world.

    For the sake of simplicity, the period of skin development can be correlated with the in utero life of the individual. This is not an absolute correspondence, however, because some of the structures of the skin may be fully formed early in the fetal period whereas other structures or regions are not complete until well into the postnatal years. Also, full establishment of adult functions of the skin always requires an extended period of development beyond the stages in utero. Development is therefore the first period in a continuum of events that modifies the skin; it is characterized by morphogenetic processes, the activation of new genes and the gain of function. In contrast, ageing may involve morpholytic processes in which genes are turned off, resulting in a loss of function. Consideration of this continuum, and the genetic and environmental interactions that come into play at progressive ages throughout life, provides a conceptual framework for discussing the place and role of the events in skin morphogenesis.

    Understanding the stages and events of normal human skin development is also important from a biomedical perspective: it allows the definition of critical periods when the skin may be more vulnerable to developmental errors; it provides an opportunity to study the evolution of skin function, establishing a background for understanding the natural history of expression of genetic skin disease in its earliest form; and it provides the essential information for the evaluation of skin samples used in the prenatal diagnosis of genodermatoses for which molecular methods are still not adaptable. Studies of skin development can also shed light on a number of basic problems in contemporary biology: epithelial–mesenchymal interactions that establish organs (in skin, these tissue interactions occur in follicle, sweat gland and nail formation); cell–cell interactions through soluble mediators; gene regulation; apoptosis; differentiation (structural, biochemical and functional); and certain longstanding basic phenomena of development such as induction, pattern formation and differentiation. They provide an opportunity to ask how a molecule is expressed and how expression of that molecule may modify the properties of the tissue over a time sequence that begins in the simple embryonic environment and builds progressively to more complex fetal conditions. Samples of fetal skin and fetal skin-derived cells are easily established in culture for experimental studies designed to probe some of these questions, and many of the studies of skin development can be performed in animal models. But the animals that are typically used for this work (primarily rodents) lack the advantage of the long gestation period of the human, which allows for the events of development to unfold slowly and thus permits a more systematic analysis of change.

    The unique morphological properties of developing human skin have always intrigued investigators who have had access to this tissue. Specific aspects of the skin that are found only in the fetus, such as the periderm, and specific events that result in the formation of complex structures, such as follicle or sweat gland, were often described for specific ages only (reviewed in refs [3–6]). These descriptions, coupled with speculation as to function, led inevitably to more systematic and comprehensive studies to characterize the complete ontogeny of the tissue, region or structure (reviewed in refs [3–6]). Such studies then began to include data derived from biochemical or immunohistocytochemical assays for the expression of specific molecules that were known to correlate with the state of differentiation or with a specific property such as barrier function adhesion and so on. Culturing and grafting human embryonic and fetal skin (reviewed in refs [7–10]) and skin-derived cells [11,12] and evaluation of skin from fetuses affected with genodermatoses (reviewed in refs [13–15]), or under conditions of growth retardation, have also provided insight into human skin development. Our understanding of skin development continues to increase as we apply more modern tools of biology to study the skin at all stages of life.

    References

    1 Lind R, Parkin FM, Cheyne GA. Biochemical and cytological changes in liquor amnii with advancing gestation. J Obstet Gynaecol Br Commonw 1971;76: 673–83.

    2 Benzie RJ, Doran TA, Harkins JL et al. Composition of the amniotic fluid and maternal serum in pregnancy. Am J Obstet Gynecol 1974;119:798–810.

    3 Holbrook KA. Structure and function of the developing human skin. In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular Biology of the Skin, 2nd edn. Oxford: Oxford University Press, 1991: 63–110.

    4 Holbrook KA. Structural and biochemical organogenesis of skin and cutaneous appendages in the fetus and neonate. In: Polin RA, Fox WW (eds) Neonatal and Fetal Medicine Physiology and Pathophysiology. New York: Grune & Stratton, 1992: 527–51.

    5 Holbrook KA, Wolff K. The structure and development of skin. In: Fitzpatrick TB, Eisen AZ, Wolff K et al. (eds) Dermatology in General Medicine, 6th edn. New York: McGraw-Hill, 1993: 97–144.

    6 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R (eds) Pediatric Dermatology, 2nd edn. New York: Churchill Livingstone, 1995.

    7 Holbrook KA, Minami SA. Hair follicle morphogenesis in the human: characterization of events in vivo and in vitro. NY Acad Sci 1991;642:167–96.

    8 Zeltinger J, Holbrook KA. A model system for long term, serum-free, suspension organ culture of human fetal tissues: experiments using digits and skin from multiple body regions. Cell Tissue Res 1997;290:51–60.

    9 Lane AT, Scott GA, Day KH. Development of human fetal skin transplanted to the nude mouse. J Invest Dermatol 1989;93:787–91.

    10 Gilhar A, Gershoni-Baruch R, Margolis A et al. Dopa reaction of fetal melanocytes before and after skin transplantation onto nude mice. Br J Dermatol 1995;133:884–9.

    11 Oliver AM. The cytokeratin expression of cultured human foetal keratinocytes. Br J Dermatol 1990;123:707–16.

    12 Scott G, Ewing J, Ryan D et al. Stem cell factor regulates human melanocyte–matrix interactions. Pigment Cell Res 1994;7:44–51.

    13 Holbrook KA, Smith LT, Elias S. Prenatal diagnosis of genetic skin disease using fetal skin biopsy samples. Arch Dermatol 1993;129:1437–54.

    14 Sybert VP, Holbrook KA, Levy M. Prenatal diagnosis of severe dermatologic diseases. Adv Dermatol 1992;7:179–209.

    15 Sybert VP, Holbrook KA. Antenatal pathology of the skin. In: Claireaux AE, Reed GB (eds) Diseases of the Fetus and Newborn: Pathology, Radiology and Genetics. New York: Cockburn, Chapman & Hall, 1995: 755–68.

    Time-Scale of Skin Development

    There are several schemes for categorizing stages of skin development (Fig. 2.1). All of them are arbitrary and overlapping, and based on some parameter that is relevant to human development per se, to a period defined for purposes of medical intervention, a specific event that is considered a landmark in the evolution of skin structure, composition and function, or on the basis of the surface properties of the developing skin [1].

    Fig. 2.1 Time-scale diagram identifying specific stages of skin development and identifying the ages at which prenatal diagnosis can be performed using each of the various methods currently employed.

    Modified from Polin RA, Fox WW. Fetal and Neonatal Physiology, 2nd edn, Vol. 1. Philadelphia: W.B. Saunders, 1998: 730.

    c02f001

    Human development is separated into embryonic (before the onset of bone marrow function) and fetal periods, corresponding, respectively, to conception until approximately 2 months estimated gestational age (EGA), and to 2 months EGA until birth. The first trimester includes the entire embryonic period and the first stages of the fetal period. Histogenesis of all skin regions is initiated in the embryo, and differentiation of some of those tissues begins to occur [2]. The boundary between the first and second trimesters, at 3 months of age, is based only on fetal age and not on any remarkable changes in structure, composition or function of any region of the skin.

    The second trimester includes many important events in skin development that can be correlated with changes in function and, at the same time that morphogenesis of new structures is initiated, there is terminal differentiation of others. During the third trimester, as far as we know, all parts of the skin are assembled and the functions of each of them are unfolding. The end of this period is not the final state of the skin, as there is significant reorganization of certain units of the skin (e.g. the vasculature), additions to the skin in volume (e.g. the dermal matrix) and functional maturation of many structures of the skin (e.g. nerves, sweat glands and stratum corneum) after the newborn faces the environment of the external world [3–6,7].

    Other important times that should be recognized in skin development are the ages at which chorionic villus sampling, amniocentesis and fetal skin biopsy are performed for the purpose of evaluating the condition of a fetus at risk for a genetic skin disease. Fetal DNA can be extracted from chorionic villi sampled around 10 weeks EGA, amniotic fluid cells can be obtained at around 14–16 weeks EGA, and fetal skin can be sampled as early as 16 weeks EGA. More typically, the skin is biopsied at 19–21 weeks [8–10]. The older age is necessary when diseases of keratinization are in question.

    This chapter is organized to describe specific periods, unique features and special events or processes of skin development using EGA, the age from the date of conception, to represent the age of the embryo or fetus in utero. In the literature gestational age is used interchangeably with menstrual age, which is calculated from the date of the last known menstrual period and thus records the timing of developmental events about 2 weeks later than EGA. Unanswered questions will be raised for the reader’s thoughtful consideration.

    References

    1 Holbrook KA, Odland GF. The fine structure of developing human epidermis: light, scanning and transmission electron microscopy of the periderm. J Invest Dermatol 1975;65:16–38.

    2 Holbrook KA, Dale BA, Smith LT et al. Markers of adult skin expressed in the skin of the first trimester fetus. Curr Prob Dermatol 1987;16:94–108.

    3 Holbrook KA. Structure and function of the developing human skin. In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular Biology of the Skin, 2nd edn. Oxford: Oxford University Press, 1991: 63–110.

    4 Holbrook KA. Structural and biochemical organogenesis of skin and cutaneous appendages in the fetus and neonate. In: Polin RA, Fox WW (eds) Neonatal and Fetal Medicine Physiology and Pathophysiology. New York: Grune & Stratton, 1992: 527–51.

    5 Holbrook KA, Wolff K. The structure and development of skin. In: Fitzpatrick TB, Eisen AZ, Wolff K et al. (eds) Dermatology in General Medicine, 6th edn. New York: McGraw-Hill, 1993: 97–144.

    6 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R (eds) Pediatric Dermatology, 2nd edn. New York: Churchill Livingstone, 1995.

    7 Holbrook KA. A histologic comparison of infant and adult skin. In: Boisits E, Maibach HI (eds) Neonatal Skin: Structure and Function. New York: Marcel Dekker, 1982: 3–31.

    8 Holbrook KA, Smith LT, Elias S. Prenatal diagnosis of genetic skin disease using fetal skin biopsy samples. Arch Dermatol 1993;129:1437–54.

    9 Sybert VP, Holbrook KA, Levy M. Prenatal diagnosis of severe dermatologic diseases. Adv Dermatol 1992;7:179–209.

    10 Sybert VP, Holbrook KA. Antenatal pathology of the skin. In: Claireaux AE, Reed GB (eds) Diseases of the Fetus and Newborn: Pathology, Radiology and Genetics. New York: Cockburn, Chapman & Hall, 1995: 755–68.

    Embryonic Skin

    The primitive ectoderm of the developing blastocyst is established at 1 week EGA, and by 20–50 days EGA the major organs and organ systems of the human embryo are becoming established. The integumentary system exhibits characteristics of the skin at 30 days EGA, the earliest age at which specimens can be realistically obtained for study. Sampling of skin at this age is remarkable considering that the 6-week-old embryo has a crown–rump length of only 20 mm – no larger than a 20 pence piece (or a dime). Nonetheless, the epidermis, dermoepidermal junction (DEJ) and dermis are well delineated and the tissue is innervated and vascularized (Fig. 2.2). The boundary between the dermis and subcutaneous tissue is not clearly defined in all body sites, but in some regions these two zones are distinct from one another on the basis of a greater density of cells and matrix in the dermis compared with the hypodermis. The skin is closely associated with the underlying developing striated muscle or cartilage on the appendages. There is no morphological evidence that epidermal appendages have begun to form.

    Fig. 2.2 (a) Tissue of the body wall of a 36-day EGA human embryo and (b) the skin from a 45-day EGA human embryo. Note the two-layered epidermis, dermis and subcutaneous tissue and the more linear orientation of dermal cells in contrast to the pleiomorphic shapes of the subcutaneous mesenchyme. In (b) note the periderm and basal cells of the epidermis, the closely associated fibroblastic cells in the dermis proximal to the epidermis and a nerve–vascular plane separating the dermis from the subcutaneous tissue (×200).

    c02f002

    In most regions of the embryo, the epidermis is a simple, flat, two-layered epithelium consisting of basal and periderm cells (see below) (Figs 2.2 and 2.3). Both types of cells are mostly filled with glycogen, a molecule that is characteristic in the cytoplasm of developing and regenerating tissues, where it most likely serves as a source of energy [1] (Fig. 2.3). The nucleus is centrally located in periderm and basal cells, and the cytoplasmic organelles are sparse and distributed either around the nucleus or at the periphery of the cell (Fig. 2.3b). The structural characteristics of these cells therefore fail to reveal significant differences between the two layers or to verify that either layer is composed of keratinocytes. Evaluation of the structural proteins, however, indicates that both contain keratin intermediate filament proteins (Fig. 2.4), but different species [2,3], and some cell-surface molecules are unique to each layer [4]. The latter markers may reflect the differences in environments surrounding each layer.

    Fig. 2.3 Transmission electron micrographs of the embryonic epidermis. In (a) note the glycogen (G)-filled basal (B) and periderm (P) layer cells. Desmosomes are evident between basal cells and between basal cells and periderm cells. The DEJ is flat and shows few sites of increased density, suggesting sites of desmosome formation. In (b) one periderm cell and portions of two basal cells are shown. Note the nature and disposition of cytoplasmic organelles within both cell types, the keratin filaments associated with desmosomes (arrow) and the microvilli extending from the periderm surface (a, ×11,525; b, ×25,000).

    c02f003

    Fig. 2.4 Immunostained samples of (a) early (∼50-day EGA) and (b–d) later (∼60-day) human embryonic epidermis showing positive staining of both periderm and basal layers with the AE1 (a) and AE3 (d) monoclonal antibodies that recognize keratins. Both layers are negative when reacted with the AE2 (c) antibody, which recognizes the differentiation-specific keratins (×350).

    c02f004

    The columnar-shaped basal cells of the embryonic epidermis express the keratins that are characteristic of adult basal layer keratinocytes – K5 (58 kDa) and K14 (50 kDa) [2,3] – and additional keratin polypeptides – K19 (40 kDa) and K8 (52 kDa) – are specific to embryonic/fetal basal cells and periderm cells [2,3]. The latter keratins are not characteristic of normal adult basal keratinocytes, but they are identified in glandular and simple epithelial cells [5]. In contrast to the adult tissue, the filaments in fetal embryonic epidermis are dispersed in the cytoplasm or assembled in small, seemingly short, bundles that are associated primarily with desmosomes and hemidesmosomes (see Fig. 2.3b). Periderm cells and basal cells also differ in the expression of many growth factors, growth factor receptors (Fig. 2.5) [6,7], cell adhesion molecules [8] and other cytoplasmic and cell-surface molecules.

    Fig. 2.5 Section of skin from a 78-day EGA human fetus showing differential expression of the A-chain of PDGF in the basal and intermediate cell layers (green) and an absence of staining in peridermal cells. The receptor for PDGFA, PDGFR-α (red), is expressed by cells in the dermis (×350).

    c02f005

    About 15–19% of the basal cells incorporate label when viable tissue is incubated in culture medium containing [³H]-thymidine for 1 h, rinsed and then processed for autoradiography. This is a higher labelling index than the fetal or adult epidermis, which was recorded at 10% and 6.7% labelling, respectively, under the same conditions [9]. It is possible that only the basal layer is truly epidermis.

    A thin, flattened layer of periderm cells covers the basal layer, with no apparent correspondence in number with the cells beneath it, i.e. one polygonal periderm cell lies above several basal cells. Microvilli project from the peridermal surface into the amniotic fluid (Figs 2.3b and 2.6). At least one keratin polypeptide expressed in periderm cells is different from those in the basal cells, K18 (45 kDa), although it is a marker for Merkel cells [10,11].

    Fig. 2.6 Scanning electron micrograph of the surface of 55-day EGA embryonic skin from the surface of the developing foot. The layer of cells shown is the periderm. Note the microvilli and the variable size and shape of the cells (×1000).

    c02f006

    Two of the immigrant cells that are prominent in adult epidermis, melanocytes (neural crest in origin) and Langerhans cells, are present in the embryonic epidermis among basal cells and associated with the basement membrane. Sheets of embryonic epidermis immunostained with an antibody that recognizes melanocytes specifically (HMB-45, an inducible, cytoplasmic antigen common to melanoma and embryonic/fetal melanocytes [12,13]) show a remarkably high density (∼1000 cells/mm²) of these cells organized in a regular pattern of distribution (Fig. 2.7). They are dendritic as early as 50 days EGA in general body skin but there is no evidence of melanosomes in the cytoplasm [14]. Langerhans cells are recognized in embryonic skin as early as 42 days EGA on the basis of a reaction product for membrane-bound Mg²+ adenosine triphosphatase (ATPase) and histocompatibility locus antigen (HLA-DR) on the plasma membrane [15–17] and by their truncated or dendritic morphology (Fig. 2.8). They are probably derived from the yolk sac or fetal liver at this age because they are present in skin before the bone marrow begins to function. At 7 weeks EGA, the density of Langerhans cells is about 50 cells/mm² [16,17].

    Fig. 2.7 Embryonic skin from a 54-day EGA human embryo immunostained with the HBM-45 monoclonal antibody, which recognizes an antigen in the melanocyte. (a) Section of skin. Note the abundance and position of these cells within the two-layered epidermis. (b) Epidermal sheet. Note the density, spacing and dendritic morphology of these cells (a, ×350; b, ×25).

    c02f007

    Fig. 2.8 Epidermal sheet from a 53-day EGA human embryo immunostained to recognize HLA-DR antigen in epidermal Langerhans cells (×400).

    Micrograph courtesy of Dr Carolyn Foster.

    c02f008

    The third immigrant cell, the Merkel cell, can be recognized in embryonic palmar skin as early as 55–60 days EGA (see Eccrine sweat gland formation) at a density of ∼130 cells/mm² [10,18], using as a marker any one of the set of keratins expressed by Merkel cells (K8, K18, K10 and K20) [10,11,19–21]. K20 is the only keratin found exclusively in Merkel cells [21]. At this embryonic age, they are distributed randomly and in a suprabasal position. Merkel cells are neuroendocrine cells that were originally thought to function primarily as slow-adapting mechanoreceptors. More recently, studies that have catalogued soluble mediators produced by these cells, for example nerve growth factor (NGF) [22], suggest that it is likely that Merkel cells are targets for ingrowing nerve fibres or other cells such as the smooth muscle cells of the arrector pili muscle [23,24]. Their presence in selected sites of developing epidermal appendages (e.g. sweat glands and hair follicles) has also been suggested to stimulate or to correlate with active proliferation of the tissue. It is generally accepted that Merkel cells are derived from keratinocytes in situ [10,19,21,25,26].

    A continuous basal lamina (lamina densa) underlies the two-layered epidermis and defines, morphologically, one structural component of the basement membrane zone [27–29]. The basal lamina is patchy, however, in regions of the body where the epidermis may be only a single layer, for example superior to the spinal cord. The molecules and antigens characteristic of all basal laminae (type IV collagen, laminin, heparan sulphate proteoglycan, nidogen/entactin) are present in the earliest recognized basal lamina of the skin; skin-specific molecules are recognized later during the first trimester in accord with the more prominent development of the attachment structures (see below) [30,31]. A thin, mat-like layer of microfilaments lies just inside the basal plasma membrane of the basal cell keratinocytes (Fig. 2.9). It may reinforce this surface of the epidermis and add to the strength of the DEJ at this stage when the structural modifications associated with dermoepidermal adhesion (hemidesmosomes, anchoring filaments, anchoring fibrils) are rudimentary [32]. The same organization of filaments is observed in cultured keratinocytes, which do not typically form hemidesmosomes and anchoring fibrils in vivo, and in basal keratinocytes under pathological situations, such as junctional epidermolysis bullosa, in which the epidermis separates from the dermis.

    Fig. 2.9 Enlarged view of the DEJ of human embryonic epidermis showing the microfilament network within the basal epidermal cell (arrows), sites where desmosomes are forming (arrowheads) and the lamina densa. Note collagen fibrils (C) surrounding the dermal fibroblastic cells (×11,625).

    c02f009

    The antigens associated with the attachment structures (laminin 5/epiligrin/kalinin and 19 DEJ-1 for hemidesmosomes and anchoring filaments [33–36]; type VII collagen for anchoring fibrils [37]) are not seen by light microscopic immunostaining methods until early in the fetal period. It is likely, however, that keratinocytes begin to synthesize these proteins in the embryonic period but that the methods used for detection are not sensitive enough to demonstrate their low levels of expression. The dermoepidermal boundary is flat in the embryonic skin (Figs 2.2, 2.3 and 2.9) and thus presents a limited surface area for nutrients to traverse between the dermis and the epidermis. This may be relatively less important in the developing skin than in infant and adult skin because the dermis is thin and the small, dispersed bundles of dermal matrix proteins and the hydrated condition of the interstitial matrix permit more rapid diffusion of substances than the mature skin.

    The dermis in the embryo is highly cellular (Figs 2.2 and 2.10), but it also contains the extracellular fibrous matrix proteins, types I, III, V and VI interstitial collagens, characteristic of adult dermis [32,38–40,41–45]. Small bundles of collagen accumulate in a thin, dense layer, called the reticular lamina, immediately beneath the dermoepidermal interface (Figs 2.2b, 2.5 and 2.9). They are also dispersed throughout the dermis in varying densities according to the collagen type and age of the embryo. Types I, III and VI collagen are distributed uniformly throughout the dermis whereas type V collagen is concentrated primarily along basement membranes (at the DEJ and around blood vessels) and surrounding cells (Fig. 2.11). Fibre bundles within the interstitial spaces are widely dispersed by a hydrated, hyaluronic acid-rich proteoglycan matrix [46–48] (Fig. 2.12). The fluidity of the matrix at this stage permits migration of mesenchymal cells to sites of active tissue morphogenesis.

    Fig. 2.10 Transmission (a) and scanning (b) electron micrographs of the embryonic dermis at 48 days EGA beginning at the DEJ. The matrix is less evident in the sectioned sample (a) than in the whole-mount specimen (b) (a, ×4500; b, ×1500).

    c02f010

    Fig. 2.11 Samples of embryonic skin immunostained with antibodies that recognize type I (a), III (b), V (c) and VI (d) collagens. Note that all of the collagens are concentrated beneath the DEJ but types III and V, especially, are found in association with all basement membranes. Types I, III and VI are found in the matrix throughout the dermal and subcutaneous tissue (a–c, ×150; d, ×300).

    Immunostaining courtesy of Dr Lynne T. Smith.

    c02f011

    Fig. 2.12 Section of the body wall from a 57-day EGA embryo treated with the Alcian blue/periodic acid–Schiff (PAS) histochemical stains. The bright pink staining of the epidermis (glycogen) and DEJ (glycoproteins) indicates a PAS-positive reaction. The blue dermis reflects the high content of hyaluronic acid. The dermal–subcutaneous boundary is marked by a transition to a lighter slightly purple reaction indicating more of the collagen–glycosaminoglycan complex (×300).

    Immunohistochemistry courtesy of Dr Richard Frederickson.

    c02f012

    A broader zone of sulphated proteoglycan-rich matrix, called the compact mesenchyme, is delineated beneath the epidermis on the basis of its rich concentration of cells that express growth factor receptors – the platelet-derived growth factor receptor β (PDGFR-β) and PDGFR-α (see Fig. 2.5), nerve growth factor receptor (NGFR) – and cell adhesion molecules (e.g. neural cell adhesion molecule, NCAM) [49,50]. Evidence from the skin of non-human species during development has shown enlargement of the composition of growth factors and receptors and adhesion molecules that are included in this dermal zone (reviewed in refs [49] and [51–53]). The compact mesenchyme may be involved in the exchange of signals between the epidermis and dermis and may be very important in stimulating the onset of appendage formation. Many of the growth factors that correspond to the receptors on the mesenchymal cells are produced by cells of the developing epidermis (e.g. PDGF-AA, PDGF-BB and NGF) (Fig. 2.5). The compact mesenchyme may also be the earliest evidence of a papillary dermis. In the adult, the modified composition and structure of the papillary dermis probably reflects molecular interactions between the epidermal and dermal cells, similar to the situation of the compact mesenchyme.

    Elastic fibres per se are not formed in the embryonic skin, but fibrillin (the microfibrils of elastic fibres) (Fig. 2.13) and elastin proteins of the elastic fibre can be identified immunohistochemically [32,38–42,44] and microfibrils can be seen by electron microscopy [32].

    Fig. 2.13 Section of skin from a 57-day EGA human embryo immunostained with an antifibrillin antibody. Note staining throughout the dermis (×200).

    Immunostaining courtesy of Dr Lynne T. Smith.

    c02f013

    Fine nerve fibres and capillaries are present within the compact mesenchyme and deeper dermis (Fig. 2.14a), and large nerve trunks and vessels are readily apparent in the subcutaneous region. Reconstructions of vessels from serial sections of developing first-trimester skin have shown that the the basic pattern of cutaneous vasculature is established in the first trimester [54]. New vessels presumably both form de novo from dermal mesenchyme and sprout from deeper, established vessels through a process that includes endothelial cell migration, capillary budding and vessel remodelling [55]. The events and mechanisms of these processes have not been explored beyond the morphological descriptions and antigenic characterization of the endothelial cells at various ages during development [55,56]. Pieces of full-thickness skin and sections of skin immunostained with an antibody that demonstrates all cutaneous nerves (protein gene product 9.5 or PGP 9.5) [57,58] reveal finely beaded nerve filaments distributed in an impressive density in the subepidermal region and in association with blood vessels (Fig. 2.14b and c). The number of fibres recognized by this antibody increases during development as the fibres become organized in networks throughout the dermis and in relation to developing epidermal appendages [59]. At 7 weeks EGA, a few calcitonin gene-related product (CGRP)-immunopositive fibres, denoting sensory fibres, are also evident [59], but autonomic nerves are not yet recognized in the skin. Staining the tissue with the p75 low-affinity NGFR antibody also reveals the patterns of nerve fibres and specific concentrations of mesenchymal cells (e.g. around developing hair follicles; see below) [60]. Both nerves and vessels are visible in stained, full-thickness samples of the nearly transparent skin (Fig. 2.15).

    Fig. 2.14 Sections of human embryonic skin at 42 days EGA (a) and 59 days EGA (b) immunostained with PGP 9.5, which recognizes all cutaneous nerves, and of a sample of 52-day EGA embryonic skin (c) immunostained with p75 antibody, which recognizes the low-affinity NGFR. Note the large nerve trunks deep in the subcutaneous tissue (a), the significant density of the fine fibres in the tangential section of the dermis of (b) and the distribution of both nerves and vessels (c) (a, ×100; b, ×200; c, ×200).

    c02f014

    Fig. 2.15 Nerves and vessels in the skin of a 79-day EGA human fetus immunostained with an anti-neurofilament

    Enjoying the preview?
    Page 1 of 1