Basic Science


Figure 1-1 The adherens junction and desmosome. (A) The adherens junction complex contains classic cadherins as transmembrane constituents, and α-catenins, β-catenins, and plakoglobin as cytoplasmic constituents. A classic cadherin is directly coupled through its cytoplasmic tail to β-catenin or plakoglobin, which in turn is linked to α-catenin, which binds to actin. (B) The desmosome complex includes desmogleins and desmocollins as transmembrane constituents, and plakoglobin, plakophilin, and desmoplakin as cytoplasmic constituents. Desmogleins and desmocollins associate with plakoglobin, which in turn binds to desmoplakin and links keratin to the membrane. N = amino-terminus; C = carboxy-terminus. (From Bolognia JL, Jorizzo JL, Rapini RP. Dermatology, 3rd Ed. Elsevier. 2012)


Cells originate in the cuboidal basal layer and flatten out as they ascend to the surface – four to five layers/strata (deep to superficial): stratum basale, s. spinosum, s. granulosum, s. lucidum (only on palmoplantar surfaces), and s. corneum


Stratum basale: mitotically active cuboidal cells from which the upper layers of the epidermis are derived


Attached to dermis by hemidesmosomes


Keratins 5 and 14 produced here


Cellular proliferation stimulated by various factors, including trauma and UV (↑ornithine decarboxylase expression is associated with (a/w) proliferative states)


Ornithine decarboxylase is inhibited by corticosteroids, retinoids, and vitamin D3


10% of cells in the basal layer are stem cells, which give rise to other stem cells and transient amplifying cells that can still replicate, but only for a few cycles, until they reach a terminal differentiation phase, where they move upwards and eventually desquamate


Transit time from basal layer to stratum corneum = 14 days; transit through the stratum corneum/desquamation = 14 days (total = 28 days from basal layer to desquamation)


Stratum spinosum: named for the “spiny” appearance of intercellular desmosomal connections on microscopy


Contain multiple types of intercellular junctions


Keratins 1 and 10 made here


Terminal keratinocyte differentiation 2° to ↑intracellular calcium in suprabasal epidermis


Odland bodies (lamellar granules) are produced by Golgi bodies in spinous layer


Primarily contain ceramide (most important lipid involved in epidermal barrier function; the most prevalent/important lipid in the stratum spinosum), along with glycoproteins, glycolipids, and phospholipids


Are specialized lysosomes that exert most of their action in the stratum corneum, by discharging ceramides and other lipids to the extracellular space of the junction between the stratum granulosum and stratum corneum ➔ ceramides help form the cornified cell envelope (see below), and eventually replace the cell membrane


Flegel’s disease and Harlequin ichthyosis are 2° to ↓lamellar granules


X-linked ichthyosis occurs due to absent steroid sulfatase in lamellar granules


Stratum granulosum: flattened cells with prominent basophilic keratohyaline granules, which contain profilaggrin (converted to filaggrin at junction of stratum granulosum and stratum corneum), loricrin, keratin intermediate filaments, and involucrin


Cells begin to lose nuclei, but keep overall structure


Cornified cell envelope production primarily takes place in the granular layer (Fig. 1-2)


Cross-linked protein and lipid structure encased in extracellular lipids forming a strong polymer that eventually replaces the plasma membrane


Process starts with envoplakin, periplakin, and involucrin scaffolding along the inner cell membrane (which is eventually replaced by ceramides from lamellar granules)


Further reinforcement by cross-linking loricrin (#1 component of cornified envelope, first appears in granular layer; mutated in Vohwinkel syndrome variant lacking deafness), small proline-rich proteins, keratin, and filaggrin


Cross-linking occurs via transglutaminase I ➔ γ-glutamyl lysine isopeptide bonds (Boards factoids: TG-1 is mutated in lamellar ichthyosis; TG-3 is antigenic target in dermatitis herpetiformis)


Other components include envoplakin (helps connect desmosomes to cornified envelope), periplakin, elafin, and others


Outer surface of the cornified envelope is ultimately surrounded by lipids (primarily ceramide) = cornified lipid envelope


Ultimately provides strong water-impermeable outer barrier


image

Figure 1-2 Formation of the cornified cell envelope (CE). Terminal differentiation of keratinocytes is triggered by an increase in the intracellular Ca2+ concentration of the suprabasal epidermis. CE assembly is initiated in the upper spinous layer via formation of a cross-linked scaffold composed of envoplakin, periplakin, and involucrin along the inner surface of the cell membrane (1). This is followed by (or perhaps coincident with) extrusion of lamellar granules into the extracellular space (2). Specialized ω-hydroxyceramides are delivered to, and eventually replace, the cell membrane, where they become linked to scaffold proteins. Reinforcement occurs via cross-linking and translocation to the cell periphery of loricrin (accounts for >80% of the mass of the CE) and small proline-rich proteins (SPRs) (3). Complexes of keratin and filaggrin also become cross-linked to the CE. In addition, proteases play important roles in processing of CE proteins and the proteolysis of corneodesmosomes that is required for desquamation. A mature and terminally differentiated cornified cell thus consists of keratin filaments covalently attached to the CE, which is composed of protein and lipid envelope components and is imbedded in the extracellular lipid lamellae. Defects in transglutaminases, lipid metabolism, CE structural proteins, and proteases leads to a variety of diseases characterized by ichthyosis and/or keratoderma (1–3). CHILD, congenital hemidysplasia with ichthyosiform erythroderma and limb defects; LI, lamellar ichthyosis; CIE, congenital ichthyosiform erythroderma. (Courtesy, Julie V Schaffer, MD) (From Bolognia JL, Jorizzo JL, Rapini RP. Dermatology, 3rd Ed. Elsevier. 2012)

Stratum corneum: outermost layer, which serves as a mechanical barrier between the epidermis and the environment


Composed primarily of protein-rich corneocytes (“bricks”; contain NO nuclei; keratin filaments attached to cornified envelope) embedded in a lipid matrix (“mortar,” cornified lipid envelope)


Serves as a barrier to water loss (conditions that perturb the skin ➔ ↑transepidermal water loss) and toxins/infectious agents


Epidermal cells of importance


Keratinocytes are the primary cells of the epidermis and produce proteins (e.g., keratin filaments) and lipids important for barrier function


Keratins: intermediate filaments that comprise the primary cytoskeleton of the epidermis (see Table 1-3)


Type I keratins: low-MW; acidic; K9-28, K31-40 (hair keratins); chromosome 17


Type II keratins: high-MW; basic; K1-8, K81-86 (hair keratins); chromosome 12


Basic structure is an α-helical rod domain (consisting of heptad amino acid repeats) divided into four segments (1A, 1B, 2A, and 2B) that are interrupted by three nonhelical segments (“linkers”)


Functional unit consists of heterodimers of type I and type II filaments that form tetramers and ultimately filaments


Anchored to plasma membrane by desmosomes


40–70 kD


Keratinocytes produce IL-1, IL-6, IL-8, IL-10, IL-12, and TNF-α, among others


Keratinocytes respond to IL-2, IL-4, IL-13, IL-22, and TNF-α, among others


Melanocytes


Neural crest-derived melanin-producing dendritic cells found in the stratum basale (≈1 : 10 ratio with keratinocytes, when viewed in 2-dimensional plane)


c-kit activation is needed for melanocyte development/migration; piebaldism occurs as a result of c-kit loss ➔ impaired melanocyte migration and proliferation; c-kit mutations are a/w mucosal and acral melanoma


Each melanocyte interfaces with 36 keratinocytes when analyzed three-dimensionally (epidermal melanin unit)


Melanin is produced in melanosomes (lysosome-type organelles) from its precursor, tyrosine, through a multistep enzymatic process involving tyrosinase (copper-dependent enzyme)


Tyrosine ➔ (tyrosinase-dependent step) DOPA ➔ (tyrosinase-dependent step) DOPAquinone ➔ pheomelanin (yellow/red; made by round melanosomes) or eumelanin (black/brown; made by elliptical melanosomes)


Melanosomes are transported along dendritic processes and transferred to keratinocytes through phagocytosis of dendrite tips


Racial variation in pigmentation: identical melanocyte density in dark and light skinned individuals; melanosomes in darker skinned individuals are larger, darker (↑melanin), more stable, and are transferred individually (vs smaller, lighter, less stable, and clustered melanosomes in lighter skin)


Melanin production is stimulated by melanocyte-stimulating hormone (MSH) and ACTH activity on MC1-R on melanocytes; also stimulated through various pathways induced by UV radiation


MC1-R loss of function mutations ➔ ↑pheomelanin:eumelanin ratio (phenotype = red hair/fair skin, ↑risk of melanoma)


Melanin absorbs UV ➔ protects against UV-induced mutations


UV exposure ➔ immediate tanning (from oxidation of existing melanin) and delayed tanning (requires new melanin synthesis)


Other high-yield examination facts:


Defects in enzymes required to convert tyrosine to melanin ➔ oculocutaneous albinism; OCA1 (Tyrosinase), OCA2 (P gene), OCA3 (TRP-1)


Defects in packaging of melanosome-specific proteins ➔ Hermansky-Pudlak syndrome (HPS1 > HPS3 > other gene mutations)


Defects in lysosome and melanosome trafficking to dendrites ➔ Griscelli (MYO5A, RAB27A, and MLPH mutations) and Chédiak-Higashi syndrome (LYST mutations)


Langerhans cells (LCs): major antigen presenting cells (APC) of the skin


Dendritic histiocytes characterized by reniform (kidney shaped) nuclei, and tennis racket-shaped Birbeck granules seen on electron microscopy


Interact with keratinocytes via E-cadherin


Positive immunostains: CD207 (langerin; most sensitive IHC stain; specific for Birbeck granules), CD1a, S100, CD34, vimentin, and actin


Originate from CD34+ progenitor cells in bone marrow like other monocytes/macrophages


Found mainly in stratum spinosum, where it first encounters and processes antigens, and subsequently migrates to the lymph nodes to activate T-cells


Downregulated in skin after UV exposure ➔ ↓immune surveillance


See p. 24 for further discussion of function


Merkel cells: slow-adapting mechanoreceptors found in fingertips, lips, oral cavity, and hair follicle ORS


Found in stratum basale; communicate with neurons


CK20+ in perinuclear dot pattern sensitive/specific for Merkel cells; also (+) for neurofilaments, S100, synaptophysin, chromogranin A, vasoactive intestinal peptide, neuron-specific enolase, and calcitonin gene-related peptide


Basement membrane zone (BMZ) (Fig. 1-3 and see Table 1-2)


Semipermeable barrier between epidermis and dermis that also serves to adhere basal keratinocytes to the underlying dermis


Key steps within each location:


Basal keratinocyte/hemidesmosome: intracellular keratin filaments (K5 and K14) attach to electron-dense hemidesmosomal plaques (plectin and BPAG1 [BP230]) on the basal plasma membrane ➔ hemidesmosomal plaque proteins bind to intracellular portions of the anchoring filaments (BPAG2 and α6β4 integrin)


Lamina lucida: extracellular portion of anchoring filaments (BPAG2, α6β4 integrin, and laminin 332) extend from the hemidesmosome down to the lamina densa; the thin filaments result in an electron-lucent region; is the weakest portion of BMZ ➔ is zone of separation in salt-split skin and also in suction blisters


Lamina densa: anchoring filaments attach to type IV collagen (#1 component) and other proteins (laminin 332, laminin 331, and nidogen) in the lamina densa ➔ results in attachment between basal keratinocyte and lamina densa


Sublamina densa: loops of type VII collagen (anchoring fibrils) arise from the underside of lamina densa, extend down into the dermis, hooking around dermal type I and III collagen fibers, and then loop back up to reattach to lamina densa (or anchoring plaques in dermis) ➔ firmly anchors the lamina densa (and all aforementioned structures) to the papillary dermis


BMZ also functions as a permeability barrier: heparan sulfate proteoglycan (negatively charged) in lamina densa is a major contributor


image

Figure 1-3 Interactions of selected molecules within the epidermal basement membrane. These interactions promote epidermal adhesion and also play a key role in a number of dermatologic diseases. Important molecular interactions include those between: (1) plakin family members, BPAG1 and plectin, with keratin intermediate filaments; (2) the former with BPAG2 and integrin α6β4 (specifically the large cytoplasmic domain of integrin subunit β4); (3) the cytoplasmic domains of BPAG2 and integrin subunit β4; (4) the extracellular domains of BPAG2 and integrin subunit α6 as well as laminin 332 (formerly laminin 5); (5) integrin α6β4 in hemidesmosomes and laminin 332 in the lamina densa; (6) laminin 332 and type VII collagen; (7) type VII collagen with type IV collagen, fibronectin, and type I collagen in the sublamina densa region. (From Bolognia JL, Jorizzo JL, Rapini RP. Dermatology, 3rd Ed. Elsevier. 2012)


Table 1-2


Basement Membrane Zone Proteins










































































Protein Site Source Family Function Disease State

BPAg1


(230 kD)

Hemidesmosome/keratinocyte Keratinocyte Plakin Binds keratins and integrins; intracellular/part of attachment plaque BP, EB simplex

BPAg2


(180 kD)


Hemidesmosome/ keratinocyte ➔ lamina lucida


Amino terminus is intracellular and carboxy terminus is extracellular


–NC16A domain is closer to amino terminus but is extracellular

Keratinocyte Collagen (XVII) Transmembrane protein and one of the anchoring filaments; interacts with BPAg1, laminin 5, β4 integrin, and plectin

N16A Terminus: BP, pemphigoid gestationis, linear IgA bullous disease


Carboxy Terminus: Cicatricial pemphigoid

α6β4 Integrin Hemidesmosome/ keratinocyte ➔ lamina lucida Keratinocyte Integrin Interacts with keratins, laminin 5, plectin, BPAg1, BPAg2; part of the anchoring filaments Ocular cicatricial pemphigoid (antibodies to β4), EB with pyloric atresia (85%)
Laminin 332 (laminin 5, epiligrin) Lamina lucida ➔ Lamina densa Keratinocyte Laminin Connects other anchoring filaments (BPAg2 and α6β4 integrin) to collagen VII; part of the anchoring filaments Antiepiligrin pemphigoid (a/w malignancy), JEB-Herlitz
Plectin Hemidesmosome Keratinocyte Plakin Binds keratins and integrins; intracellular/part of attachment plaque EB with muscular dystrophy, EB with pyloric atresia (15%)
Nidogen (entactin) Lamina densa Unclear Nidogen Adaptor between laminin 1 and collagen IV in lamina densa; stabilizes proteins of lamina densa
Collagen IV Lamina densa Unclear Collagen Anchors laminins in lamina densa ➔ structural support; also a component of anchoring plaques in dermis, which attach collagen VII to collagen I and III Goodpasture disease, Alport syndrome
Collagen VII Sublamina densa Fibroblasts Collagen Major component of anchoring fibrils Dystrophic EB, bullous lupus, EB acquisita
Heparan sulfate proteoglycan Lamina densa Fibroblasts Proteoglycans Contribute to matrix of and give an overall negative charge (creating a permeability barrier) to the basement membrane


Table 1-3


Protein Components of the Epidermis (Including Nonepidermal Keratins)




























































































































Protein Site of Synthesis Function Disease State
Keratin 1 Suprabasal keratinocytes (produced in spinous layer) Primary keratinocyte cytoskeleton Epidermolytic ichthyosis (preferred new name for EHK), epidermolytic and nonepidermolytic (Unna-Thost) PPK, ichthyosis hystrix of Curth-Macklin*
Keratin 2 Granular layer Superficial epidermolytic ichthyosis (Siemens)
Keratin 3 Cornea Meesmann’s corneal dystrophy
Keratin 4 Mucosal epithelium White sponge nevus
Keratin 5 Basal keratinocytes EBS, Dowling-Degos disease*
Keratin 6a Outer root sheath of hair Pachyonychia congenita I*
Keratin 6b Nail bed epithelium Pachyonychia congenita II
Keratin 9 Palmoplantar suprabasal keratinocytes Vorner (epidermolytic) PPK
Keratin 10 Suprabasal keratinocytes (produced in spinous layer) Epidermolytic ichthyosis*
Keratin 11 Granular layer
Keratin 12 Cornea Meesmann’s corneal dystrophy
Keratin 13 Mucosal epithelium White sponge nevus
Keratin 14 Basal keratinocytes EBS, Naegeli-Franceschetti-Jadassohn syndrome, dermatopathia pigmentosa reticularis
Keratin 16 Outer root sheath of hair Pachyonychia congenita I*
Keratin 17 Nail bed epithelium Pachyonychia congenita II, steatocystoma multiplex
Keratin 19 Stem cells of basal layer
Keratin 71, 73, 74 Hair inner root sheath Wooly hair
Keratin 32, 35, 82, 85 Hair cuticle
Keratin 17, 33, 34, 36, 37, 75, 81 Hair medulla Pseudofolliculitis barbae
Keratins 31–38, 81, 83, 85, 86 Hair cortex Monilethrix (KRT81, KRT83, KRT86 most commonly; also DSG4)
Filaggrin/profilaggrin Granular layer

Aggregates keratin, flattening granular layer cells. Degraded in the stratum corneum into urocanic acid and pyrrolidone carboxylic acid, which help block/absorb UV radiation.


Urocanic acid is also a component of natural moisturization factor – helps keep stratum corneum hydrated/moist

Ichthyosis vulgaris, atopic dermatitis
Loricrin Granular layer Most abundant component of cornified cell envelope. Cross-linked to involucrin by transglutaminase 1**.

Vohwinkel syndrome with ichthyosis (NO deafness)


Decreased in psoriasis

Involucrin Granular layer Component of cornified cell envelope. Proteins are cross-linked together by transglutaminase 1 ➔ strong border Increased in psoriasis


*In psoriasis and other hyperproliferative states, keratin 6 and 16 are upregulated and keratin 1 and 10 are downregulated



**Transglutaminase 1 mutations ➔ lamellar ichthyosis and NBCIE


Dermis


Located below the epidermis, derived from mesoderm, and divided into papillary dermis (superficial) and reticular dermis (deep)


Cells of significance


Fibroblasts-create extracellular matrix and are involved in wound healing


Mononuclear phagocytes – discussed on p. 23


Mast cells – discussed on p. 23


Glomus cells – specialized smooth muscle cells derived from Sucquet-Hoyer canals, which allow for blood shunting from arterioles to venules (bypassing capillaries); found mainly in the palms/soles


Overproduction ➔ glomus tumor (favors acral sites because of ↑glomus cell density)


Dermal dendritic cells – bone marrow-derived APC that resides within dermis; highly phagocytic


Extracellular matrix (ECM)


Provides structure and support to the dermis; essential for water retention and for signal transduction


Synthesized by dermal fibroblasts


Composed of collagens, elastin, fibrillins, fibulins, integrins, laminins, glycoproteins, and proteoglycans


Collagens are triple helices formed by amino acid chains where every third residue is glycine (Gly-X-Y), with a high likelihood of proline and hydroxyproline/hydroxylysine in the X and Y positions, respectively


Accounts for 75% of dry weight of the skin; #1 component of the dermis


Collagen I is the primary collagen (85%) of the ECM; type III (10%; important and prevalent in blood vessels, fetal skin, GI tract, new scars, and keloids) and V are also present


Lysyl hydroxylase and proline hydroxylase catalyze crosslinking of collagen; vitamin C-dependent process (deficiency ➔ scurvy)


Defects in collagen and/or collagen cross-linking result in most forms of Ehlers-Danlos syndrome: COL1A1/2 (EDS arthrochalasia type, and osteogenesis imperfecta); COL3A1 (EDS vascular type); COL5A1/2 (classical EDS); Lysyl hydroxylase/PLOD1 gene (EDS kyphoscoliosis type)


Matrix metalloproteinases degrade collagen


Retinoids ➔ ↑collagen production


Corticosteroids and UV ➔ ↓collagen production


Elastic fibers provide resilience from stretching and modulate TGF-β and BMP signaling


Account for 4% of dry skin weight


90% elastin (core) and 10% fibrillin (surrounds elastin); elastin contains high levels of desmosine and isodesmosine ➔ these crosslink with fibrillin via lysyl oxidase (copper necessary for function)


Notable defects in elastic tissue: Fibrillin 1 mutation (Marfan’s syndrome); Fibrillin 2 mutation (Congenital contractural arachnodactyly); Fibulin 5 (Cutis laxa; gene defect results in decreased desmosine); LEMD3 mutation (Buschke-Ollendorf syndrome; defect results in increased desmosine); ABCC6 mutation (Pseudoxanthoma elasticum; mutation results in fragmentation and calcification of elastic fibers)


Elaunin fibers run horizontal/parallel in reticular dermis and oxytalan fibers run vertical/perpendicular to DEJ in papillary dermis; mnemonic: “stand (= vertical) up-high (= high in dermis) with OXYgen (= OXYtalan)”


UV radiation ➔ damage of elastic fibers


All aforementioned fibers are set in a matrix of proteoglycans and glycosaminoglycans (GAGs) that retain large quantities of water (up to 1000× their volume!) = ground substance


Most important GAGs = hyaluronic acid, chondroitin sulfate, dermatan sulfate, and heparan sulfate


GAGs are broken down by lysosomal hydrolases


Cutaneous vasculature


Cutaneous vasculature important for metabolic support of the skin and maintenance of body temperature


Two vascular plexuses: superficial (➔ vessels of the reticular dermis) and deep (➔ follicles, glands)


VEGF is the primary mediator of vasculogenesis


↑VEGF: most cancers, psoriasis, POEMS syndrome, and other diseases with increased vasculature


Lymphatic vessels collect fluid and proteins from interstitium and direct it into the lymph circulation


Prox1, Podoplanin (D2-40), LYVE-1, and VEGFR-3 are lymphatic vessel markers


Cutaneous neurology


Nerves of the skin are responsible for detecting touch/pressure, pain, itch, and other sensations


Cutaneous sensory nerves are divided into free nerve endings and corpuscular nerve endings (round/globular collection of neural and other cells)


Free nerve endings


Itch and pain: A-δ (larger; myelinated) and C-polymodal nociceptor afferent fibers (smaller; unmyelinated)


End in the epidermis/superficial dermis


Specialized nerve receptors (corpuscular nerve endings)


Krause end bulbs: genitalia, perianal region, and vermillion lips; mnemonic “Krazy Krause ends on erotic areas”


Meissner’s corpuscle: superficial (dermal papillae) mechanoreceptor of digits; fast adapting; suited for pressure/light touch


Pacinian corpuscle: deep (deep dermis/fat) mechanoreceptor of palmoplantar skin, nipples, and genital region; fast adapting; suited for vibration and deeper pressure


Merkel nerve ending: superficial (basal epidermis) mechanoreceptor most concentrated in fingertips, lips, and external genitalia; slow adapting; suited for pressure/touch


Ruffini corpuscle: deep (fat) mechanoreceptor most concentrated around fingernails; slow adapting; suited for sustained pressure


Innervation of cutaneous appendages:


Adrenergic control: vascular smooth muscle, apocrine glands, and arrector pili contraction


Cholinergic control: eccrine glands


Adnexal structures


Eccrine glands


Secretory exocrine gland primarily responsible for thermoregulation and waste excretion


Found on all cutaneous surfaces except: external auditory canal, lips, glans penis, clitoris, and labia minora


Highest concentration = palms and soles


Controlled by hypothalamus; innervated by postganglionic sympathetic fibers, which synapse with muscarinic acetylcholine receptors on the glands


Isotonic sweat secreted in secretory gland ➔ NaCl reabsorbed in duct ➔ hypotonic sweat is delivered to surface


↑rate of sweating ➔ more isotonic solution (less time to reabsorb in duct)


Maximal rate of sweating ~ 3 L/hr


Merocrine secretion


Components (deep to superficial): secretory coil (deep dermis), intradermal/straight duct (eosinophilic cuticle seen on histology), and acrosyringium (intraepidermal portion; spiral duct that opens onto the skin surface)


Stains for S100, keratin, and CEA


Apocrine glands


Secretory exocrine glands with unclear function in humans, though in animals they mediate sexual attraction through pheromone release


Activity begins around puberty


Located primarily in anogenital skin, axillae, external ear canal, vermillion border, periumbilical region, eyelid margin, and breast


Empty into follicular infundibulum (above sebaceous duct)


Secretory control unclear ➔ glands noninnervated, but do have β-adrenergic receptors, which are likely stimulated by circulating catecholamines


Secretory products released through decapitation secretion: cholesterol and cholesterol esters, triglycerides, squalene, and fatty acids


Lipofuscin = pigmented mixture of lipids and proteins ➔ responsible for yellow-brown color of chromhidrosis


Initially odorless secretions ➔ later modified by surface bacteria ➔ results in body odor


Ectopic or modified apocrine glands: mammary glands, ceruminous glands of the external auditory canal, and Moll’s gland of the eyelids


These empty directly to the surface


Sebaceous glands


Secretory exocrine glands found primarily on the scalp, face, and upper anterior trunk (“seborrheic areas”)


NOT on the palms/soles


Functions include water retention and innate immune defense


Consist of sebocytes, which contain lipid vacuoles


Normally associated with hair follicles and empty into inferior portion of the infundibulum


Pubertal androgen production is major signal for sebaceous gland maturation (under adrenergic control)


Transient maternal androgen stimulation present in infancy


Other endocrine factors stimulating maturation and sebum production: MSH, CRH, and substance P


Secretory products released through holocrine secretion (entire cell lyses to release contents):


Triglycerides (#1 component; ≈50%) > wax esters (#2) > squalene (#3)


Others: cholesterol esters, cholesterol, antimicrobial peptides, androgens, and cytokines


Ectopic sebaceous glands: Meibomian glands on eyelid tarsal plate, Fordyce spots (vermillion lip/oral mucosa), Montgomery tubercles (areolae/nipples), Tysons glands (labia minora/prepuce), and Zeis glands (eyelid margin, close to Moll’s gland)


Hair


Epithelial-derived appendage important for temperature regulation, protection of other structures (nasal mucosa, eyes, and ears), social and sexual cues, and tactile sensory input


Three types of hairs:


Lanugo – fine hairs shed late in gestation and during the first month of life


Vellus – fine hairs over face, trunk, and extremities early in life


Terminal – coarse, darker hairs of scalp, eyebrows, and eyelashes; postpubertal androgens induce switch to terminal hairs in other sites


Hair density: ~100,000 hairs on scalp; more in blonde and fewer in red-haired individuals


Anatomy: Table 1-4



Table 1-4


Hair Anatomy

















































Portion of Hair Description
Hair bulb Lowermost portion of the hair follicle
Hair matrix Rapidly proliferating keratinocytes that terminally differentiate to produce the hair shaft
Infundibulum Region extending from the skin surface down to the point where the sebaceous gland opens into the hair follicle; ORS displays cornification similar to that of the interfollicular epidermis (i.e., contains keratohyaline granules)
Isthmus Region located between the opening of the sebaceous gland, down to the site of insertion of the arrector pili muscle; ORS displays trichilemmal keratinization (no inner root sheath and IRS is shed before this point)
Lower hair follicle Region located between hair bulb to proximal isthmus; encapsulates dermal papilla; has inner and outer root sheaths; critical line of Auber is the widest area
Arrector pili muscle Inserts at the level of the bulge; pulls up hair (“goose bumps”)
Bulge Segment of the outer root sheath located at the level of arrector pili muscle insertion; major seat of epithelial stem cells of the hair follicle
Secondary hair germ Additional seat of epithelial and also of melanocyte stem cells; located between club hair and dermal papilla in telogen hair follicle
Connective tissue sheath (CTS) Special mesenchymal follicular sheath that is tightly attached to the hair follicle basement membrane and is continuous with the follicular dermal papilla
Follicular dermal papilla (DP) Onion-shaped, closely packed, specialized fibroblast population with inductive and morphogenic properties; hair cycle-dependent fibroblast trafficking occurs between CTS and DP; volume of DP determines size of hair bulb and, thus, hair shaft diameter
Inner root sheath (IRS) Packages and guides the hair shaft; cornifies normally; stains red secondary to citrulline; not present in telogen hairs; is present in lower hair follicle but not in the isthmus/infundibulum
Outer root sheath (ORS) Merges distally into the epidermis and proximally into the hair bulb; provides slippage plane, nutrition, regulatory molecules, and stem cells
Critical line of Auber Widest section of the hair bulb and where most mitotic activity essential for hair growth occurs
Follicle pigmentary unit Melanin-producing hair follicle melanocytes located up and around the upper one-third of the DP; transfer pheomelanosomes or eumelanosomes to differentiating hair follicle keratinocytes in the precortical matrix; goes largely into apoptosis during each catagen phase, regenerated from melanocyte stem cells in hair germ during anagen

(Adapted from Bolognia JL, Jorizzo JL, Rapini RP. Dermatology, 3rd Ed. Elsevier. 2012)


Follicular layers (outer to inner): glassy membrane, outer root sheath, inner root sheath (Henle’s layer, Huxley’s layer, and cuticle), and hair shaft (cuticle, cortex [where most hair keratins located], and medulla)


Cuticle helps keeps hair intact – damage ➔ split ends (trichoptilosis)


Cuticle from hair shaft and inner root sheath merge


Dermal papilla: mesenchymal structure (from embryonic mesoderm) containing vasculature; contributes to hair cycle regulation


Hair cycle: Table 1-5


Exogen: phase of active shedding of club hair between telogen and anagen


Lose about 100 hairs/day


Kenogen: subphase of telogen in which no shaft is present



Table 1-5


Hair Growth Cycle
























Anagen Catagen Telogen (Club Hairs)
Phase activity Growth (~0.4 mm/day or ~1 cm/month on scalp); follicular melanocytes only active in anagen phase) Regression (melanocytes in matrix apoptose; inner root sheath lost) Resting
Duration 2–6 years 2–3 weeks 3 months
Percentage (%) of scalp hairs 85–90 1–2 10–15

Color of hair 2° to hair melanocytes in anagen bulb/matrix (melanin unit = 1 melanocyte: five keratinocytes; melanocytes only produce pigment in anagen phase!)


Eumelanin (brown/black hair pigment) vs pheomelanin (red/blonde hair pigment)


Hair follicle stem cells reside in bulge and contribute to hair cycling, tissue regeneration, and wound healing; follicular melanocytes can migrate to interfollicular areas in disease states (e.g., vitiligo) to assist with repigmentation


As with interfollicular stem cells in the basal layer of skin (see p. 1), also produce transient amplifying cells with restricted mitotic capacity


Disulfide bonding via cysteine residues determines curliness of hair – these bonds are broken when hair is straightened, but subsequently reform with time


Nail


Appendageal structure important for protection and function of fingertips, for proper function of the feet, ability to scratch, and aesthetic appearance


Plate is composed of keratin-producing onychocytes


Important anatomic structures: Table 1-6



Table 1-6


Nail Anatomy


































Nail Location Description
Proximal nail fold Superficial layer continuous with skin, deep layer continuous with nail matrix
Eponychium (cuticle) Located between nail plate and nail matrix; acts as seal against the environment
Nail matrix At the proximal end of nail unit, generates the plate. Proximal matrix➔ superficial portion of nail plate; distal matrix ➔ ventral portion of plate. Melanocytes found in nail matrix
Lunula (distal nail matrix) Junction between matrix and bed
Nail plate Hard, functional unit of nail, composed primarily of keratins; strong attachment to nail bed
Nail bed Extends from lunula to onychodermal band. Provides support for nail plate. Very minimal contribution to nail plate synthesis
Onychodermal band Red/pink transverse band marking end of bed
Hyponychium Continuous with ventral edge of free nail plate and distal fingertip skin
Lateral nail folds Guide growth of nail plate

Growth rate


Fingernails: 2 to 3 mm/month (~6 months to grow out)


Toenails: 1 mm/month (~12 months to grow out)





1.2 Embryology



Skin structures derived from two of three primary germ layers


Ectoderm: epidermis, adnexal structures, Merkel cells, melanocytes (neural crest), and nerves (neuroectoderm)


Mesoderm: fibroblasts, LCs, vessels, and inflammatory cells


Epidermis


5 weeks: outer periderm and inner basal epidermal layer


8 weeks: epidermal stratification


9–12 weeks: melanocytes, LCs, and Merkel cells migrate into the epidermis


Late second trimester: terminal differentiation, with full stratification


Basement membrane, dermis, and subcutis


6–8 weeks: fibroblasts appear beneath the epidermis


9 weeks: distinct border between epidermis and dermis (DEJ present)


9–12 weeks: primordial vasculature formed


16–18 weeks: initial fat formation in subcutis


20 weeks: mature thickness of dermis and dermal ridges present


Hair


9–12 weeks: initial follicle development on the eyebrows, scalp, upper lip, and chin – spreads caudally and ventrally; epidermal placodes (derived from ectoderm) induce underlying dermal papilla formation (derived from mesoderm)


18–20 weeks: hair canal fully formed


24–28 weeks: initiation of cycling through anagen, catagen, and telogen


Sonic hedgehog is an important molecule for telogen to anagen transition


Nails


8–10 weeks: nail bed demarcation


12 weeks: proximal nail folds formed


17 weeks: nail plate formed, covers nail bed by week 20


Adnexal glands


10 weeks: eccrine gland anlage formation on palms and soles


14–16 weeks: eccrine primordia bud down, glands begin to develop


22 weeks: initiation of truncal eccrine gland formation; eccrine glands and ducts nearly mature on volar skin


Apocrine gland formation initiated later than eccrine glands, at 22 weeks


Sebaceous gland formation parallels hair follicle development (derived from outer root sheath)


Melanocytes


Neural crest-derived cells, migrate under the direction of KIT and KIT ligand


12 weeks: melanocytes present in epidermis


12–16 weeks: melanin production begins


16–20 weeks: melanocytes proliferate and become fully functional (transfer melanosomes to keratinocytes)


Skin stem cell biology


Epidermal stem cells responsible for maintenance, repair, and renewal of epidermis


Keratinocyte stem cells located within the bulge region of the hair follicle and at the base of rete ridges of interfollicular epidermis


Complete renewal of epidermis every 40 to 56 weeks


Stem cells are multipotent with unlimited capacity to divide


Asymmetric division gives rise to transient amplifying cells, which divide rapidly to produce terminally differentiated cells

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

May 4, 2017 | Posted by in Dermatology | Comments Off on Basic Science

Full access? Get Clinical Tree

Get Clinical Tree app for offline access