Skin and Adnexal Structures


Inflammatory Conditions

Superficial Perivascular Dermatitis

Dermatitis With Minimal Epidermal Changes

Superficial Dermatophytosis (Tinea)

Clinical Features

  • Caused by three genera of imperfect fungi— Epidermophyton, Trichophyton, and Microsporum —that cause superficial infections involving keratinized tissues such as the cornified layer of epidermis, the hair, and the nails

  • Dermatophytosis involving different anatomic sites named with site-specific terms (scalp-tinea capitis, beard-area-tinea barbae, face-tinea faciei, trunk-tinea corporis, intertriginous areas-tinea cruris, feet and hands-tinea pedis et manus, and nails-tinea unguium)

  • Typical lesions present as sharply demarcated annular patches and plaques with an arcuate border

  • Tinea capitis and barbae present as folliculitis; tinea unguium is characterized by yellow-gray discoloration of nails

Histopathology

  • Parakeratosis with neutrophils, mild/minimal spongiosis ( Figure 2.1A )

    Figure 2.1, Dermatophytosis.

  • Mild/minimal, superficial perivascular lymphocytic infiltrate

  • Fungi present as filamentous hyphae, spores, or yeast forms in the cornified layer and in the hair shafts in cases of tinea capitis and tinea barbae (endothrix infection—dermatophytes within the hair shaft, ectothrix infection—dermatophytes confined to the surface of the hair shaft)

Special Stains and Immunohistochemistry

  • Periodic acid–Schiff (PAS) reaction stains fungi deep red to pink, and Gomori methenamine silver (GMS) stains fungi black ( Figure 2.1B )

Other Techniques for Diagnosis

  • Microbiologic cultures useful in speciation of the fungal organisms

  • Polymerase chain reaction (PCR) techniques particularly helpful for detecting dermatophyte infections of the nail (onychomycosis)

Differential Diagnosis

  • Vitiligo

    • Decreased density/complete of absence of basal melanocytes

  • Urticaria

    • Presence of dermal edema, telangiectases, and eosinophils

  • Candidiasis

    • Seen in patients with impaired host response, especially patients with hematologic malignancies and diabetes

    • Can present as cutaneous, mucocutaneous, or systemic infection

    • Presence of spongiotic or subcorneal pustules in which pseudohyphae can be demonstrated with PAS/GMS stain

  • Pityriasis (Tinea) Versicolor Caused by Genus Malassezia

    • Affects upper trunk with brownish discoloration that may become hypopigmented

    • Presence of round spores and thick, short hyphae (“spaghetti and meatballs”) recognizable as faintly basophilic, refractive structures in routine hematoxylin and eosin (H&E)–stained sections diagnostic

Pearls

  • Identification of fungal organisms on routine H&E-stained sections aided by lowering the microscope condenser, which enhances the refractile nature of the fungi

  • Fungi in the cornified layer are sandwiched between a lower zone of parakeratosis and an upper zone of orthokeratosis (“sandwich sign”)

  • The presence of neutrophils in a slightly parakeratotic cornified layer and mild superficial perivascular dermatitis should always prompt a PAS stain in search of fungal elements

  • Deep folliculitis with intense acute and granulomatous inflammation due to dermatophyte infection is known as “Majocchi granuloma”

Selected References

  • Eldridge M.L., Chambers C.J., Sharon V.R., Thompson G.R.: Fungal infections of the skin and nail: new treatment options. Expert Rev Anti Infect Ther 2014; 12: pp. 1389-1405.
  • Gottlieb G.J., Ackerman A.B.: The “sandwich sign” of dermatophytosis. Am J Dermatopathol 1996; 8: pp. 347-350.
  • Kondori N., Tehrani P.A., Strömbeck L., Faergemann J.: Comparison of dermatophyte PCR kit with conventional methods for detection of dermatophytes in skin specimens. Mycopathologia 2013; 176: pp. 237-241.

Vitiligo

Clinical Features

  • Acquired, possibly autoimmune disease with a strong familial association

  • Characterized by patches of pigment loss (“hypopigmentation”) in skin

  • Localized disease may show linear, segmental pattern; generalized disease involves face, upper trunk, dorsa of hands, periorificial areas, and genitalia; scalp and eyelashes not typically affected

  • Stable patches of vitiligo are sharply demarcated and may be surrounded by a zone of hyperpigmentation; in active lesions, areas of total depigmentation may be surrounded by a zone of partial depigmentation and have a slight rim of erythema at the border

Histopathology

  • Sparse/mild superficial perivascular inflammation with scattered melanophages

  • Complete loss of melanin from the basal layer and total absence of melanocytes only seen in well-established lesions and in the depigmented center of expanding lesions

  • Advancing border-enlarged melanocytes with increased dendritic processes and a mild superficial perivascular inflammation

  • Normal-appearing skin (adjacent to the vitiliginous patches)—patchy interface change and a mild superficial perivascular and lichenoid lymphocytic infiltrate

  • Active lesions—lymphocyte aggregates (“Pautrier-like” microabscesses) in the basal layer

Special Stains and Immunohistochemistry

  • Silver stains or the dopa reaction (Fontana-Masson) highlight loss of basal layer melanin pigment ( Figures 2.2A and B )

    Figure 2.2, A, Vitiligo. Fontana-Masson stain shows loss of pigmentation at the basal cell layer. B, Normal skin. Fontana-Masson stain shows normal pigmentation at the basal cell layer.

  • Immunohistochemical stains for SOX10 or Melan-A are useful in confirming decreased density/absence of basal melanocytes

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • On routine H&E-stained sections, other diseases manifesting with minimal histologic alterations (normal-appearing skin), such as tinea versicolor, urticaria, and urticaria pigmentosa (macular variant), should be considered.

  • Mycosis fungoides-epidermotropism and Pautrier microabscesses in all epidermal layers composed of lymphocytes with hyperchromatic and convoluted nuclei diagnostic

Pearls

  • Biopsy of uninvolved/normal skin in addition to that of lesional skin (for comparative purposes) is essential for a diagnosis of early/evolving lesions.

  • Vitiligo can be seen in association with other autoimmune disorders such as thyroid disorders, pernicious anemia, and alopecia areata.

Selected References

Attili VR, Attili SK. Lichenoid inflammation in vitiligo: a clinical and histopathologic review of 210 cases. Int J Dermatol . 2008;47:663–669.

Le Poole IC, Das PK. Microscopic changes in vitiligo. Clin Dermatol . 1997;15:863–873.

Silverberg JI, Silverberg NB. Clinical features of vitiligo associated with comorbid autoimmune disease: a prospective survey. J Am Acad Dermatol . 2013;69:824–826.

Urticaria

Clinical Features

  • Typically, presents with pruritic, raised, erythematous, and edematous areas ( wheals or hives )

  • Acute urticaria lasts <6 weeks, more common in the pediatric age group; chronic urticaria lasts >6 weeks, more common in adults

  • An underlying predisposing condition can be identified in only up to 25% of patients; certain foods, drugs, contact allergens, and physical stimuli such as pressure, cold temperature, and occult infections may be factors

  • Urticarial vasculitis is a syndrome consisting of recurrent urticaria, arthralgia, and abdominal pain; individual cutaneous lesions persist for >24 hours

  • In angioedema, dermal edema extends into subcutaneous fat and presents with large wheals

Histopathology

  • Interstitial edema, telangiectases, and a sparse/mild superficial perivascular mixed inflammatory cell infiltrate composed of lymphocytes, eosinophils, and neutrophils ( Figure 2.3 )

    Figure 2.3, Urticaria.

  • Urticarial vasculitis—features of early leukocytoclastic vasculitis characterized by mild perivascular infiltrate of neutrophils, neutrophilic nuclear dust, and extravasated red blood cells; minimal or absent fibrin deposits in the vessel walls

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Hypocomplementemia, seen in 32% of patients with urticarial vasculitis; measurements of CH50 and C1q binding assays are helpful

  • Patients with hereditary angioedema have a low serum level of C1 esterase inhibitor during the attack

  • Direct immunofluorescence (DIF) studies: vascular deposits of immunoglobulins, complement, or fibrin are seen in one third of patients with urticarial vasculitis

Differential Diagnosis

  • Macular Variant of Urticaria Pigmentosa (Telangiectasia Macularis Eruptiva Perstans)

    • Generally occurs as an extensive eruption of brownish-red macules

    • Dilated blood vessels in the upper dermis and a mild superficial perivascular mononuclear cell infiltrate composed mostly of mast cells are characteristic( Figure 2.4A )

      Figure 2.4, Urticaria pigmentosa, macular type.

    • Eosinophils are generally absent; dermal edema is not prominent

    • Giemsa, toluidine blue, Leder, or immunohistochemical stain for mast cell tryptase and CD117 can help demonstrate the increased number of mast cells ( Figure 2.4B )

  • Leukocytoclastic Vasculitis Due to Other Causes

    • Fibrinoid necrosis of vessels, leukocytoclasia, and an abundance of extravasated erythrocytes

Pearls

  • Fibrinoid necrosis and thrombosis are not typically seen in urticarial vasculitis

  • In hereditary angioedema, a form of dominantly inherited angioedema, recurrent attacks of edema involve skin and oral, laryngeal, and gastrointestinal mucosa; death due to laryngeal edema can occur if not treated

  • Urticarial vasculitis may be associated with infectious mononucleosis, infectious hepatitis, and autoimmune diseases such as systemic lupus erythematosus (SLE)

Selected References

Barzilai A, Sagi L, Baum S, Trau H, et al. The histopathology of urticaria revisited-clinical pathological study. Am J Dermatopathol . 2017;39:753–759.

Cugno M, Castelli R, Cicardi M. Angioedema due to acquired C1-inhibitor deficiency: a bridging condition between autoimmunity and lymphoproliferation. Autoimmun Rev . 2008;8:156–159.

Wisnieski JJ. Urticarial vasculitis. Curr Opin Rheumatol . 2000;12:24–31.

Interface Dermatitis

Lichen Planus

Clinical Features

  • Disorder of unknown etiology involving skin, mucous membranes, hair, and nails

  • Typically presents as pruritic, polygonal, violaceous, flat-topped papules with a fine scale

  • Predilection for flexor surfaces of extremities, lower back, and glans penis

  • Surface of lesions may show a network of white lines ( Wickham striae )

  • Oral lesions (lacy, reticular network of papules involving buccal mucosa or the tongue) may be the sole manifestation

  • Association with hepatitis C

Histopathology

  • Compact hyperkeratosis, irregular epidermal hyperplasia (“saw-toothed”) with wedge-shaped hypergranulosis that corresponds to the openings of follicles and acrosyringia ( Figure 2.5A )

    Figure 2.5, A, Lichen planus. Hyperkeratosis, hypergranulosis, irregular epidermal hyperplasia, and a bandlike, predominantly lymphocytic infiltrate that obscures the dermoepidermal junction where there is also clefting (Caspary-Joseph space). Melanophages are present in the dermal infiltrate. B, Lichen nitidus. Lichenoid lymphohistiocytic infiltrate with admixed multinucleate giant cells that is confined to widened dermal papillae enclosed by elongate rete on either side (“ball and claw”).

  • Bandlike, predominantly lymphocytic infiltrate in the superficial dermis that obscures the dermoepidermal junction

  • Eosinophilic colloid bodies (“Civatte bodies”), representing damage to the basal cell layer, are present at the dermoepidermal junction

  • Small clefts known as Max-Joseph/Caspary-Joseph spaces may be seen at the dermo-epidermal junction

  • Hypertrophic lichen planus—typically characterized by irregular acanthosis and interface change with an underlying inflammatory infiltrate confined only to the tips of the rete ridges

  • Oral lesions—typically show parakeratosis, less acanthosis, and are frequently ulcerated

  • Lichen planus of hair follicles (lichen planopilaris)—active lesions typically show a dense lymphocytic infiltrate surrounding the infundibulum of the follicle ; in later stages, there is perifollicular fibrosis with loss of follicles with advanced stages resulting in scarring alopecia

Special Stains and Immunohistochemistry

  • PAS stain confirms interface change

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Lichenoid Drug Eruption

    • Focal parakeratosis and necrotic keratinocytes particularly at and above the dermoepidermal junction

    • Presence of eosinophils in the inflammatory cell infiltrate

  • Lichen Planus–Like Keratosis (Benign Lichenoid Keratosis)

    • Solitary lesion

    • Absence of epidermal hyperplasia with hypergranulosis and presence of parakeratosis in addition to lichenoid pattern of inflammation

    • Adjacent areas may show changes of solar lentigo

  • Lichenoid Graft-Versus-Host Disease (GVHD)

    • Generally the inflammatory cell infiltrate is sparse

    • Foci of parakeratosis and thinning of epidermis may be present

  • Lichen Striatus

    • More common in children than adults and presents as a unilateral eruption along Blaschko lines on extremities, trunk, or neck

    • Lichenoid inflammatory cell infiltrate similar to lichen planus; however, the infiltrate extends deep in the reticular dermis around hair follicles and sweat glands

    • Epidermal spongiosis and an admixture of histiocytes in the inflammatory cell infiltrate

  • Lichen Nitidus

    • Asymptomatic dermatosis of childhood, characterized by round, flat-topped papules that measure only a few millimeters

    • Histologically, the inflammatory cell infiltrate is bandlike but the lesion is small and discrete; infiltrate is confined to widened dermal papillae and enclosed by elongated rete on either side (“ball and claw”) ( Figure 2.5B )

    • Frequent histiocytes in the infiltrate, epidermal atrophy, and focal parakeratosis are helpful in differentiating lichen nitidus from lichen planus

Pearls

  • Parakeratosis and eosinophils in the inflammatory infiltrate are not features of cutaneous lichen planus, and indicative of other causes of lichenoid dermatitis such as lichenoid drug eruption

Selected References

Shai A, Halevy S. Lichen planus and lichen planus-like eruptions: pathogenesis and associated diseases. Int J Dermatol . 1992;31:379–384.

Shiohara T. The lichenoid tissue reaction: an immunological perspective. Am J Dermatopathol . 1998;10:252–256.

Tziotzios C, Lee JYW, Brier T, et al. Lichen planus and lichenoid dermatoses: clinical overview and molecular basis. J Am Acad Dermatol . 2018;79(5):789–804.

Erythema Multiforme

Clinical Features

  • Acute cytotoxic cell-mediated hypersensitivity reaction to infections, most commonly herpes simplex virus infection, and drugs, in particular sulfonamides

  • The eruption is multiform and consists of macules, papules, vesicles, and occasionally large flaccid bullae; often associated with fever

  • Herpesvirus-associated erythema multiforme (EM) involves the extremities and presents with typical target-like lesions, whereas drug-associated EM shows truncal involvement and a purpuric type of macular eruption; mucosal involvement (Stevens-Johnson syndrome) is characteristic

  • The most severe form, toxic epidermal necrolysis (a widespread blotchy erythema) is characterized by large flaccid bullae with “sloughing” of the skin and is most often caused by drugs, including sulfonamides, β-lactam antibiotics, and nonsteroidal anti-inflammatory drugs; associated with a high mortality rate

Histopathology

  • Cornified layer is unaltered, attesting to the acute nature of the disease

  • Confluent interface change/vacuolar alteration of the basal cell layer and a sparse superficial perivascular lymphocytic infiltrate that may focally obscure the dermoepidermal junction ( Figure 2.6A )

    Figure 2.6, A, Erythema multiforme. Vacuolar alteration of the basal cell layer above which there are necrotic keratinocytes. B, Toxic epidermal necrolysis. Full-thickness epidermal necrosis with separation at the dermoepidermal junction. The cornified layer is unaltered, attesting to the acute nature of the process, and there is only a minimal inflammatory cell infiltrate.

  • The hallmark of EM is the presence of necrotic keratinocytes, initially as single cells and later as small clusters; the necrosis is more widespread in drug-induced EM

  • In toxic epidermal necrolysis, there is full-thickness epidermal necrosis resulting in subepidermal bullae ( Figure 2.6B )

  • In late lesions, the papillary dermis may contain melanophages (a sign of damage to the basal cell layer)

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • DIF studies of lesional skin <48 hours old will show immunoglobulin M (IgM) and C3 in the walls of superficial dermal vessels

  • Herpes simplex virus DNA has been detected within lesions of EM using PCR and in situ hybridization (ISH)

Differential Diagnosis

  • Staphylococcal Scalded-Skin Syndrome

    • Clinical appearance may be similar to toxic epidermal necrolysis

    • Microscopically, staphylococcal scalded-skin syndrome shows a split in the granular layer diagnostic (in contrast to toxic epidermal necrolysis where there is separation at the dermoepidermal junction)

  • Acute GVHD

    • May be histopathologically indistinguishable from early EM

  • Fixed Drug Eruption

    • Interface dermatitis with necrotic keratinocytes similar to EM

    • Deeper extension of inflammatory cell infiltrate containing eosinophils

    • Prominent pigment incontinence

Pearls

  • Erythema multiforme, Stevens-Johnson syndrome, and toxic epidermal necrolysis are best regarded as a spectrum of the same disease process.

Selected References

Ackerman AB, Ragaz A. Erythema multiforme. Am J Dermatopathol . 1985;7:133–139.

Roujeau JC. Stevens-Johnson syndrome and toxic epidermal necrolysis are severity variants of the same disease which differs from erythema multiforme. J Dermatol . 1997;24:726–729.

Samim F, Auluck A, Zed C, Williams PM. Erythema multiforme: a review of epidemiology, pathogenesis, clinical features, and treatment. Dent Clin North Am . 2013;57(4):583–596.

Graft-versus-host disease

Clinical Features

  • Occurs most frequently in bone marrow transplant recipients and less commonly in recipients of blood products and solid organ transplants

  • Incidence is 30% to 40% in related donor-recipient versus 60% to 80% when unrelated; risk increases with age

  • Acute phase

    • Presents with the triad of skin lesions, hepatic dysfunction, and diarrhea; develops within 3 months after transplant

    • Skin lesions are characterized by extensive erythematous macules, purpuric to violaceous papules and plaques, and, in severe cases, toxic epidermal necrolysis–like eruption; oral lesions may be present

  • Chronic phase

    • Occurs >3 months to a year after transplantation

    • In the early lichenoid stage, the eruption is similar to lichen planus

    • Late sclerotic stage is characterized by skin induration and tightening

Histopathology

  • Acute phase—4 grades

    • Grade I: vacuolar alteration of the basal cell layer, which may be focal or diffuse

    • Grade II: intraepidermal necrotic keratinocytes occasionally surrounded by lymphocytes (satellite necrosis) ( Figure 2.7 )

      Figure 2.7, Acute graft-versus-host disease.

    • Grade III: widespread necrosis of keratinocytes with separation at the dermoepidermal junction

    • Grade IV: full-thickness necrosis and loss of epidermis

    • Sparse superficial perivascular lymphocytic infiltrate is usually present in acute GVHD

    • Follicular papules when evident clinically exhibit histopathologic changes in follicular epithelium similar to those evident in the epidermis

  • Chronic phase

    • Early lichenoid phase-histopathologic features of lichen planus; satellite necrosis may still be seen in GVHD

    • Late sclerotic phase-histopathologic changes similar to scleroderma with dermal sclerosis extending into the subcutis plus epidermal atrophy

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Erythema Multiforme

    • May be histopathologically indistinguishable from acute GVHD

  • Lichen Planus

    • May be histopathologically indistinguishable from lichenoid phase of GVHD

  • Scleroderma

    • May be histopathologically indistinguishable from sclerotic phase of GVHD

    • Epidermal atrophy, if present, helps in differentiating sclerotic phase of GVHD from scleroderma

Pearls

  • Histopathologic findings are phase dependent

  • Acute phase characterized by an interface (vacuolar type) dermatitis caused by the attack of donor immunocompetent T lymphocytes against histocompatibility antigens exposed on recipient cells

  • Chronic phase—initially characterized by an interface (lichenoid type) dermatitis and later as a sclerosing dermatitis and caused by immunocompetent lymphocytes that differentiate in the recipient

  • Target cells in GVHD are the stem cells in the regenerating compartment—that is, the basal keratinocytes in skin and the epithelial cells at the base of the crypts in the gastrointestinal tract

Selected References

Aractingi S, Chosidow O. Cutaneous graft-versus-host disease. Arch Dermatol . 1998;134:602–612.

Hillen U, Hausermann P, Massi D, et al. Consensus on performing skin biopsies, laboratory workup, evaluation of tissue samples and reporting of the results in patients with suspected cutaneous graft-versus-host disease. J Eur Acad Dermatol Venereol . 2015;29:948–954.

Zhou Y, Barnett MJ, Rivers JK. Clinical significance of skin biopsies in the diagnosis and management of graft-vs-host disease in early post-allogeneic bone marrow transplantation. Arch Dermatol . 2000;136:717–721.

Cutaneous Lupus Erythematosus

Clinical Features

  • Chronic multisystem autoimmune disease that affects the connective tissue and vasculature of various organs

  • Three classic cutaneous forms: discoid, subacute, and systemic lupus

    • Discoid lupus erythematosus (DLE)—clinically, mildly scaling, erythematous, edematous, sharply demarcated plaques measuring up to 15 cm, involving the scalp, face, upper trunk, and upper extremities (photosensitive distribution); follicular plugging may be seen

    • Older lesions—atrophic with variable pigmentation

      • Subacute lupus erythematosus—presents as a symmetric photodistributed eruption of psoriasiform/annular plaques

      • Tumid lupus—indurated plaques and nodules without overlying erythema or atrophy

      • Verrucous lesions—seen in 2% of patients with chronic cutaneous lupus erythematosus

      • Panniculitis—seen in select patients with chronic cutaneous or systemic forms of lupus erythematosus

Histopathology

  • DLE—epidermal atrophy, hyperkeratosis with follicular plugging and confluent interface change that involves follicular epithelia and marked thickening of the basement membrane ( Figure 2.8A )

    Figure 2.8, Cutaneous lupus erythematosus.

  • A moderate perivascular and periadnexal lymphocytic infiltrate and a mild increase in dermal mucin

  • Subacute cutaneous lupus erythematosus (SCLE) and neonatal lupus erythematosus-prominent interface change with numerous individually necrotic dyskeratotic keratinocytes, less prominent hyperkeratosis and inflammatory cell infiltrate than DLE

  • SLE—may show interface change with involvement of follicular epithelia, periadnexal and perivascular lymphocytic infiltrate, and increased dermal mucin similar to discoid lupus but more often the histologic changes are subtle

  • Tumid lupus erythematosus—superficial and deep perivascular and periadnexal lymphocytic infiltrate and increased dermal mucin but no epidermal changes

  • Lupus panniculitis/lupus profundus—lobular lymphocytic panniculitis with hyaline fat necrosis, superficial and deep perivascular and periadnexal lymphocytic infiltrate, interstitial mucin, with or without epidermal changes ( Figure 2.8B ), reactive lymphoid follicles not uncommon in the subcutis

  • Verrucous lupus—verrucous epidermal hyperplasia, interface change involving follicular epithelia, superficial and deep perivascular and periadnexal lymphocytic infiltrate and increased dermal mucin

Special Stains and Immunohistochemistry

  • PAS stain—helpful in demonstrating interface change and thickened basement membrane ( Figure 2.8C )

  • Colloidal iron stain—highlights interstitial mucin deposits

Other Techniques for Diagnosis

  • DIF—continuous granular deposition of immunoglobulin G (IgG), IgM, and C3 in a band along the dermoepidermal junction ( Figure 2.8D )

Differential Diagnosis

  • Dermatomyositis

    • Interface change, pauci-inflammatory, minimal increase in dermal mucin, and lupus band test is generally negative

  • Lichen Planus

    • Irregular epidermal hyperplasia with hypergranulosis, and lichenoid lymphoid cell infiltrate with pigment incontinence

  • Polymorphous Light Eruption

    • Superficial and deep perivascular lymphocytic infiltrates of lupus (especially tumid form) must be differentiated from polymorphous light eruption, which usually shows marked edema of papillary dermis

  • Subcutaneous Panniculitis-Like T-Cell Lymphoma

    • Can mimic lupus profundus

    • Rimming of the subcutaneous fat cells by neoplastic T lymphocytes and presence of lymphocyte nuclei within the cytoplasm of histiocytes (emperipolesis) are characteristic features

Pearls

  • Interface dermatitis with involvement of follicular epithelia diagnostic for all forms of lupus erythematosus

  • Increase in interstitial mucin is minimal in SCLE and DLE

  • Site of biopsy crucial for DIF-exposed normal skin for SLE and lesion >3 months duration for DLE

  • Well-defined lesions of DLE occur in 15% of patients with subacute lupus erythematosus

Selected References

Arps DP, Patel RM. Lupus profundus (panniculitis): a potential mimic of subcutaneous panniculitis-like T-cell lymphoma. Arch Pathol Lab Med . 2013;137:1211–1215.

Biazar C, Sigges J, Patsinakidis N, Ruland V, et al. Cutaneous lupus erythematosus: first multicenter database analysis of 1002 patients from the European Society of Cutaneous Lupus Erythematosus (EUSCLE). Autoimmun Rev . 2013;12:444–454.

Jerdan MS, Hood AF, Moore GW, et al. Histopathologic comparison of the subsets of lupus erythematosus. Arch Dermatol . 1990;126:52–55.

Dermatomyositis

Clinical Features

  • Connective tissue disease characterized by inflammatory myositis involving the proximal muscles and cutaneous lesions consisting of a heliotrope rash (violaceous, slightly edematous periorbital patches involving the eyelids), Gottron papules (discrete red-purple papules over bony prominences of knuckles, knees, and elbows), and photodistributed erythematous-edematous lesions over the upper chest and back

  • The disease has two peaks, one in childhood and one between the ages of 45 and 65 years

Histopathology

  • Histopathologic changes of the erythematous-edematous lesions of the skin include epidermal atrophy, vacuolar interface change with scattered individually necrotic keratinocytes, a sparse superficial perivascular lymphocytic infiltrate ( Figure 2.9 )

    Figure 2.9, Dermatomyositis.

  • Mild increase in interstitial mucin

  • Subepidermal fibrin deposits can be seen

  • Gottron papules—epidermal hyperplasia in addition to mild interface change

  • Panniculitis and calcification of the subcutaneous tissue may be seen at a later stage

Special Stains and Immunohistochemistry

  • PAS stain confirms interface change and a thickened basement membrane zone

Other Techniques for Diagnosis

  • DIF—may show complement (C5b-C9) around blood vessels

Differential Diagnosis

  • Subacute Cutaneous Lupus Erythematosus

    • Histopathologic features identical to those of dermatomyositis, clinical correlation including evaluation for myositis critical

    • Positive lupus band test helpful (especially in early stages of dermatomyositis when the muscular weakness is not apparent)

Pearls

  • Pauci-inflammatory interface dermatitis with a minimal increase in dermal mucin

  • Histopathologic reaction pattern that of a subtle interface dermatitis and is therefore not diagnostic

  • Clinicopathologic correlation key to making the histopathologic diagnosis

Selected References

Callen JP. Dermatomyositis. Lancet . 2000;355:53–57.

Mendese G, Mahalingam M. Histopathology of Gottron’s papules-utility in diagnosing dermatomyositis. J Cutan Pathol . 2007;34:793–798.

Sontheimer RD. Cutaneous features of classic dermatomyositis and amyopathic dermatomyositis. Curr Opin Rheumatol . 1999;11:475–482.

Epidermal Spongiosis

Spongiotic Dermatitis

Clinical Features

    • Spongiotic dermatitis refers to reaction pattern characterized histologically by the presence of intercellular edema (spongiosis) in the epidermis

    • Group includes allergic contact dermatitis, photoallergic dermatitis, nummular dermatitis, atopic dermatitis, dyshidrotic dermatitis, and Id reaction

  • Allergic Contact Dermatitis

    • Most commonly caused by poison ivy, nickel, and rubber compounds, presents with pruritic, edematous, erythematous papules and occasional vesicles usually within 1-3 days of exposure

  • Photoallergic Dermatitis

    • Due to topical application (photocontact) or ingestion of an allergen

    • Presents with pruritic and erythematous papulovesicular lesions on sun-exposed skin; usually on face, arms, and neck

  • Nummular Dermatitis

    • Disease of unknown etiology characterized by coin-shaped, pruritic, erythematous, scaly, crusted plaques on the exterior aspects of extremities

  • Atopic Dermatitis

    • Inherited chronic, pruritic, scaly eruption affecting face and extensor aspects of extremities in children

  • Dyshidrotic Dermatitis

    • Characterized by numerous pruritic vesicles along sides of fingers and toes and palms and soles

  • Autoeczematization or Id Reaction

    • Sudden localized or generalized eruption of pinhead-sized vesicles developing in association with a defined local dermatitis or with infection

    • Most common cause is a remote dermatophyte infection

Histopathology

  • Three distinct reaction patterns— acute , subacute , and chronic

  • Acute spongiotic dermatitis

    • Spongiotic vesicles with minimal acanthosis ( Figure 2.10A )

      Figure 2.10, Spongiotic dermatitis.

    • In allergic contact dermatitis, eosinophils may be present in the vesicles ( Figure 2.10B )

    • Mild papillary dermal edema and a superficial perivascular lymphohistiocytic inflammation are present

  • Subacute spongiotic dermatitis

    • Parakeratosis with plasma, mild to moderate spongiosis, epidermal hyperplasia, and superficial perivascular lymphohistiocytic infiltrate

  • Chronic Spongiotic Dermatitis

    • Hyperkeratosis, acanthosis with hypergranulosis, minimal spongiosis, and papillary dermal fibrosis

Special Stains and Immunohistochemistry

  • PAS stain useful in excluding dermatophytosis with spongiosis

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Seborrheic Dermatitis

    • Histopathology varies with age of the lesion, follicle-centered, “shoulder” parakeratosis ( Figure 2.10C ) and a mild superficial perivascular lymphoid cell infiltrate with admixed neutrophils

  • Pityriasis Rosea

    • “Moundlike” parakeratosis ( Figure 2.10D ), mild spongiosis ( only in the “herald patch” ), mild superficial perivascular lymphoid cell infiltrate, and extravasated red cells

    • Identical changes may be seen in superficial form of erythema annulare centrifugum

  • Spongiotic Drug Eruptions and Insect Bite Reactions

    • Deeper extension of the inflammatory infiltrate with admixed eosinophils

  • Psoriasis

    • Chronic spongiotic dermatitis (lichen simplex chronicus) may resemble psoriasis but lacks confluent parakeratosis with neutrophils and thinning of suprapapillary plates

Pearls

  • Eczema is a nonspecific term used clinically to describe erythematous vesicular lesions with scaly crust that show spongiotic dermatitis on histology

  • Three distinct reaction patterns—acute, subacute, and chronic

  • Histopathology typically varies with age of the lesion

  • Several entities can exhibit the same reaction pattern

Selected References

Ackerman AB, Chongchitnant N, Sanchez J, et al. Histologic Diagnosis of Inflammatory Skin Diseases . Baltimore: Williams & Wilkins; 1997.

Ackerman AB, Ragaz A. A plea to expunge the word “eczema” from the lexicon of dermatology and dermatopathology. Am J Dermatopathol . 1982;4:315.

Weedon D. Skin Pathology . 2nd ed. New York: Churchill Livingstone; 2002:112.

Incontinentia Pigmenti

Clinical Features

  • An X-linked–dominant dermatosis that affects mostly females

  • Characteristic cutaneous manifestations seen at birth include crops of vesicles and bullae on extremities arranged in a linear or whorled pattern

  • Lesions heal with hyperkeratosis and verrucous epidermal hyperplasia; the verrucous lesions heal with streaks and whorls of hyperpigmentation that are later replaced by faint hypochromic patches

Histopathology

  • Vesicular stage—characterized by marked epidermal spongiosis with eosinophils, individually dyskeratotic keratinocytes, and whorls of squamous cells with central keratinization ( Figure 2.11 )

    Figure 2.11, Incontinentia pigmenti.

  • Verrucous stage—characterized by hyperkeratosis, verrucous epidermal hyperplasia, individually dyskeratotic keratinocytes, and occasional eosinophils

  • Hyperpigmented stage—characterized by numerous melanophages in the superficial dermis

  • Hypopigmented stage—characterized by epidermal atrophy, decreased melanin in the basal cell layer, and absence of adnexae

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Eosinophilic Spongiosis

    • Seen in a spectrum of unrelated entities that include allergic contact dermatitis, precursor lesions of pemphigus and bullous pemphigoid; clinical history essential in differentiation

  • Epidermal Nevus

    • Verrucous stage of incontinentia pigmenti may resemble epidermal nevus

  • Postinflammatory Pigmentary Alterations

    • Hyper- and hypopigmented stages of incontinentia pigmenti resemble postinflammatory pigmentary changes

Pearls

  • Histopathology typically varies with age of the lesion with four distinct histopathologic reaction patterns

  • Several entities can exhibit the same reaction pattern, clinicopathologic correlation essential in making a conclusive diagnosis

  • Eosinophilic chemotactic activity has been shown in the blister fluid of patients with incontinentia pigmenti

  • In up to 80% of patients, systemic findings with involvement of the central nervous system and eye may be seen; teeth abnormalities may be present

  • Extent of systemic involvement determines the clinical course

  • The genetic defect in the familial form has been traced to a mutation in the IKBKG- gene (called NEMO) localized to Xq28 region

Selected References

Ackerman AB, Chongchitnant N, Sanchez J, et al. Histologic Diagnosis of Inflammatory Skin Diseases . Baltimore: Williams & Wilkins; 1997.

Berlin AL, Paller AS, Chan LS. Incontinentia pigmenti: a review and update on the molecular basis of pathophysiology. J Am Acad Dermatol . 2002;47:169–187.

Ehrenreich M, Tarlow MM, Godlewska-Janusz E, Schwartz RA. Incontinentia pigmenti (Bloch-Sulzberger syndrome): a systemic disorder. Cutis . 2007;79:355–362.

Psoriasiform Dermatitis

Psoriasis

Clinical Features

  • Chronic dermatosis of unknown etiology affecting up to 2% of the population

  • Males and females affected equally

  • Variably sized, well-demarcated plaques covered by thick, silvery white scale that have a predilection for areas with trauma, including scalp, lumbosacral skin, and extensor surfaces of elbows and knees

  • Several manifestations that include localized or generalized pustular psoriasis, eruptive or guttate psoriasis, and erythrodermic psoriasis

  • Involvement of nails, oral mucosa, and tongue can occur

Histopathology

  • Confluent parakeratosis with neutrophils and neutrophilic microabscesses (intracorneal-Munro microabscesses and intraspinous-spongiform pustule of Kogoj)

  • Hypogranulosis corresponding to zones of parakeratosis

  • Regular (psoriasiform) epidermal hyperplasia with elongation of rete ridges and thinning of suprapapillary plates ( Figure 2.12A )

    Figure 2.12, A, Psoriasis. Confluent parakeratosis with collections of neutrophils, diminished granular layer, regular (psoriasiform) epidermal hyperplasia with thinning of suprapapillary plates, dilated blood vessels in the papillary dermis, and mild superficial perivascular inflammation are seen. B, Pustular psoriasis. Prominent Munro microabscess. C, AIDS-associated psoriasis. Scale crust with neutrophils, psoriasiform epidermal hyperplasia, absence of thinning of the suprapapillary plate [a], and numerous plasma cells [b].

  • Dilated tortuous blood vessels in the dermal papillae

  • Mild superficial perivascular lymphocytic infiltrate

  • Pustular psoriasis—Exhibits the above but has in addition prominent Munro microabscesses and spongiform pustules of Kogoj ( Figure 2.12B )

  • Guttate psoriasis—psoriasiform epidermal hyperplasia not as pronounced

  • Erythrodermic psoriasis-histopathologic changes may be nonspecific

  • AIDS-associated psoriasiform dermatitis

  • Distinguishing features from psoriasis include individually necrotic/dyskeratotic keratinocytes, absence of thinning of the suprapapillary plates, and presence of plasma cells ( Figure 2.12C-a,b )

Special Stains and Immunohistochemistry

  • PAS stain helpful in excluding dermatophytic infections (which also exhibit neutrophils in the stratum corneum)

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Subacute/Chronic Spongiotic Dermatitis Such As Allergic Contact or Nummular Dermatitis

    • Presence of spongiosis and eosinophils helpful in differentiation

  • Dermatophyte Infection

    • Presence of superficial fungal spores and hyphal forms diagnostic (typically evident in PAS stained sections)

  • Bacterial Impetigo

    • Subcorneal pustule diagnostic of an early lesion (rarely biopsied)

    • Gram stain may demonstrate the bacteria

  • Pityriasis Rubra Pilaris

    • Alternating ortho and para-keratotic scale, psoriasiform epidermal hyperplasia, follicular plugging with “shoulder” parakeratosis diagnostic

    • Contrasting features with psoriasis include absence of neutrophils in the stratum corneum and retention of the granular cell layer

Pearls

  • “Dry” horn typical-scale crust with neutrophils but no inspissated serum

  • Intraepidermal pustule not specific for psoriasis—can be seen in Reiter syndrome, AIDS-associated psoriasiform dermatitis, acrodermatitis enteropathica, and glucagonoma syndrome

  • Basal keratinocyte atypia not uncommon and related to etiopathogenesis

  • Psoriatic arthritis is seen in about 15% of patients with psoriasis and characteristically involves terminal interphalangeal joints

  • Severe expression of psoriasis is seen in patients with AIDS who may present with extensive erythroderma, inverse psoriasis, and palmoplantar psoriasis

Selected References

De Mozzi P, Johnston GA, Alexandroff AB. Psoriasis. An evidence-based update. Report of the 9th evidence-based update meeting, 12 May 2011, Loughborough, UK. Br J Dermatol . 2012;166:252–260.

Nestle FO. Psoriasis. Curr Dir Autoimmun . 2008;10:65–75.

Ragaz A, Ackerman AB. Evolution, maturation, and regression of lesions of psoriasis. Am J Dermatopathol . 1979;1:199.

Pityriasis Rubra Pilaris

Clinical Features

  • Chronic follicular-based erythematous papular eruption of unknown etiology that progresses to form orange-red scaly plaques containing islands of normal-appearing skin

  • Generalized erythroderma may occur with progression

  • Palmoplantar keratoderma and scales on the face and scalp not uncommon

Histopathology

  • Alternating orthokeratosis and parakeratosis in horizontal and vertical directions (“checkerboard pattern”) ( Figure 2.13A and B )

    Figure 2.13, Pityriasis rubra pilaris.

  • Psoriasiform epidermal hyperplasia with short rete and thick suprapapillary plates

  • Follicular papules demonstrate “shoulder” parakeratosis and dilated follicular infundibula with follicular plugging

  • Acantholysis restricted to adnexal epithelium

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Psoriasis

    • Confluent parakeratosis with neutrophils and neutrophilic microabscesses, psoriasiform epidermal hyperplasia with hypogranulosis ( granular cell layer is retained in pityriasis rubra pilaris ) and basal keratinocyte atypia, thinning of suprapapillary plates ( suprapapillary plates are thick in pityriasis rubra pilaris ), and papillary dermal telangiectases are diagnostic features

Pearls

  • “Checkerboard” cornified layer distinctive

  • Acantholysis not uncommon but restricted to adnexal epithelium when present

  • “Shoulder” parakeratosis not specific to pityriasis rubra pilaris and can also be seen in seborrheic dermatitis

Selected References

Barr RJ, Young EM Jr. Psoriasiform and related papulosquamous disorders. J Cutan Pathol . 1985;12:412–425.

Mobini N, Toussaint S, Kamino H. Noninfectious erythematous, papular, and squamous diseases. In: Elder DE, Elenitsas R, Johnson BL Jr, et al., eds. Lever’s Histopathology of Skin . 10th ed. Philadelphia; Lippincott Williams & Wilkins; 2008:169.

Piamphongsant T, Akaraphant R. Pityriasis rubra pilaris: a new proposed classification. Clin Exp Dermatol . 1994;19:134–138.

Superficial and Deep Perivascular Dermatitis

Dermatitis With Minimal Epidermal Changes

Lymphocytes Predominant

Polymorphous Light Eruption

Clinical Features

  • Recurrent pruritic papules and plaques that typically occur in young women mostly during summer, induced by ultraviolet radiation (UVR)

  • Eruption starts a few minutes to a few hours after exposure and lasts for hours to days

  • Extensor aspects of arms and upper chest are most frequently involved

  • Despite the diverse clinical presentation , within any one patient, only one clinical form is consistently manifested

Histopathology

  • Epidermis is mostly unremarkable

  • Prominent papillary dermal edema with occasional subepidermal separation and extravasated red cells

  • Superficial and deep perivascular, predominantly lymphocytic infiltrate ( Figure 2.14A )

    Figure 2.14, A, Polymorphous light eruption. A superficial and deep perivascular lymphocytic infiltrate is associated with marked papillary dermal edema. B, Pernio. Interface change, superficial and deep dermal perivascular and periadnexal inflammation, increased dermal mucin, prominent papillary dermal edema, and extravasated erythrocytes.

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Sweet Syndrome

    • Superficial and deep perivascular and interstitial predominantly neutrophilic infiltrate with abundant leukocytoclasia, prominent papillary dermal edema, and extravasated erythrocytes

  • Sweet Syndrome-Like Arthropod Bite Reaction

    • Superficial and deep dermal inflammation with numerous eosinophils and prominent papillary dermal edema

  • Pernio

    • Interface change, superficial and deep dermal perivascular and periadnexal inflammation, increased dermal mucin, prominent papillary dermal edema and extravasated erythrocytes ( Figure 2.14B )

  • Pityriasis Lichenoides, Acute Form

    • Parakeratosis with neutrophils, interface change with numerous individually necrotic keratinocytes, papillary dermal edema and extravasated erythrocytes

  • Cutaneous Lupus Erythematosus

    • Typically the subacute and tumid forms should be considered in the differential diagnosis

    • Polymorphous light eruption lacks changes at the dermoepidermal junction, has less prominent periadnexal infiltrate, and lacks interstitial mucin deposits, features typically seen in cutaneous lupus

    • Papillary dermal edema is more prominent in polymorphous light eruption than in SCLE

Pearls

  • Extravasated erythrocytes and prominent papillary dermal edema helpful histopathologic features but not specific for polymorphous light eruption

  • Extravasated erythrocytes and papillary dermal edema, also observed in pityriasis lichenoides acuta, pernio, Sweet syndrome

Selected References

  • Boonstra H.E., van Weelden H., Toonstra J., van Vloten W.A.: Polymorphous light eruption: a clinical, photobiologic, and follow-up study of 110 patients. J Am Acad Dermatol 2000; 42: pp. 199-207.
  • Gruber-Wackernagel A., Byrne S.N., Wolf P.: Polymorphous light eruption: clinic aspects and pathogenesis. Dermatol Clin 2014; 32: pp. 315-334.
  • Hasan T., Ranki A., Jansen C.T., Karvonen J.: Disease associations in polymorphous light eruption: a long-term follow-up study of 94patients. Arch Dermatol 1998; 134: pp. 1081-1085.

Eosinophils Predominant

Insect Bite Reaction (Papular Urticaria)

Clinical Features

  • Allergic reaction induced by bites from mosquitoes, fleas, and bedbugs

  • Papules and papulovesicular lesions that are intensely pruritic and often excoriated

Histopathology

  • Focal epidermal ulceration/excoriation, epidermis, and cornified layer may show changes of excoriation

  • A superficial and deep, V- or wedge-shaped, perivascular and interstitial, mixed inflammatory cell infiltrate containing numerous eosinophils characteristic ( Figure 2.15A and B )

    Figure 2.15, Insect bite reaction.

  • Chronic lesions—characterized by an exuberant inflammatory infiltrate with reactive follicles

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Immunohistochemistry for B- and T-cell lymphoid markers helpful in confirming a reactive process in chronic lesions

Differential Diagnosis

  • Eosinophilic Cellulitis/Wells Syndrome

    • Pandermal, predominantly eosinophil-rich inflammatory infiltrate with “flame figures” (eosinophilic debris as a result of degranulating eosinophils) ( Figure 2.15C )

  • Dermal Hypersensitivity Reaction

    • Hypersensitivity reaction due to drugs while histopathologically indistinguishable from papular urticaria often tends to have an inflammatory infiltrate that is less dense and deep

  • B-Cell Lymphoproliferative Disease/Follicle Center Lymphoma

    • Poorly defined follicles with aberrant expression of lineage-specific immunohistochemical markers

Pearls

  • Presence of a deep, predominantly eosinophil-rich inflammatory infiltrate helpful but not diagnostic

  • Identification of foreign body material reminiscent of arthropod parts superficially or within the dermis diagnostic

  • Reactive lymphoid follicles may be a feature of chronic lesions (persistent arthropod bite reaction)

Selected References

Ackerman AB, Chongchitnant N, Sanchez J, et al. Histologic Diagnosis of Inflammatory Skin Diseases: An Algorithmic Method Based on Pattern Analysis . 2nd ed. Baltimore: Williams & Wilkins; 1997:202.

Gilliam AC, Wood GS. Cutaneous lymphoid hyperplasias. Semin Cutan Med Surg . 2000;19:133–141.

Howard R, Frieden IJ. Papular urticaria in children. Pediatr Dermatol . 1996;13:246–249.

Interface Dermatitis

Pityriasis Lichenoides

Clinical Features

  • Self-limited cutaneous eruption of unknown cause that affects young adults and children

  • Two forms:

    • Acute or pityriasis lichenoides et varioliformis acuta (PLEVA), more severe form common in children and young adults

    • Chronic or pityriasis lichenoides chronica (PLC), the milder form more common in adults

    • Transitional forms with changes between the two extremes occur

  • In PLEVA, a papular, papulonecrotic, and occasionally vesiculopustular eruption occurs on the trunk and proximal extremities, usually resolves in a few weeks; crops of new lesions can continue to appear, and the disease process itself may have a chronic course

  • In PLC, recurrent crops of reddish-brown papules with adherent scales occur on the trunk and extremities and resolve in a few weeks

Histopathology

  • PLEVA

    • Parakeratosis and scale crust with neutrophils, vacuolar interface change, and individually necrotic keratinocytes ( Figure 2.16A )

      Figure 2.16, A, Pityriasis lichenoides acuta. Parakeratosis containing collections of neutrophils, vacuolar alteration of the basal cell layer, and patchy lichenoid and perivascular lymphocytic inflammation are seen. Scattered necrotic keratinocytes and extravasated red cells are present. B, Pityriasis lichenoides chronica. Hyperkeratosis, parakeratosis, mild epidermal hyperplasia, interface change, and a superficial perivascular lymphohistiocytic infiltrate with extravasated erythrocytes

    • Superficial and deep perivascular and lichenoid, predominantly lymphocytic infiltrate with atypical CD30+ lymphocytes

    • Lymphocytic vasculitis (especially in the ulceronecrotic variant)

    • Papillary dermal edema and extravasated erythrocytes

  • PLC

    • Hyperkeratosis, parakeratosis, mild epidermal hyperplasia, interface change

    • Superficial perivascular lymphohistiocytic infiltrate with extravasated erythrocytes ( Figure 2.16B )

Special Stains and Immunohistochemistry

  • Lymphocytes predominantly of the cytotoxic/suppressor T-cell phenotype, presence of CD8+ T lymphocytes confirmed by immunohistochemistry

  • Immunohistochemical stain for CD30 may reveal scattered CD30+ cells in PLEVA

Other Techniques for Diagnosis

  • DIF may reveal IgM and C3 deposits along the basement membrane zone

Differential Diagnosis

  • Lymphomatoid Papulosis

    • Histopathologic features overlap with PLEVA

    • Wedge-shaped inflammatory cell infiltrate composed of clusters of atypical CD30+ lymphocytes with admixed eosinophils

  • Vesicular Insect Bite Reactions

    • Superficial and deep dermal infiltrate with numerous eosinophils, papillary dermal edema and extravasated erythrocytes helpful histopathologic clues

Pearls

  • Density of infiltrate in PLC is less than that observed in PLEVA

  • Despite the unifying presence of CD30+ lymphocytes in PLEVA and lymphomatoid papulosis they are pathogenetically distinct entities

Selected References

Bowers S, Warshaw EM. Pityriasis lichenoides and its subtypes. J Am Acad Dermatol . 2006;55:557–572.

Ersoy-Evans S., Greco MF, Mancini AJ, et al. Pityriasis lichenoides in childhood: a retrospective review of 124 patients. J Am Acad Dermatol . 2007;56:205–210.

Kempf W, Kazakov DV, Palmedo G, et al. Pityriasis lichenoides et varioliformis acuta with numerous CD30(+) cells: a variant mimicking lymphomatoid papulosis and other cutaneous lymphomas. A clinicopathologic, immunohistochemical, and molecular biological study of 13 cases. Am J Surg Pathol . 2012;36:1021–1029.

Fixed Drug Eruption

Clinical Features

  • Well-defined, circumscribed patches occur at the same site in response to repeated intake of the drug

  • Lesions are slightly edematous and erythematous and may develop dusky centers and become bullous and heal with pigmentation

  • Increasing number of lesions can occur with each successive administration of the offending drug

Histopathology

  • Interface dermatitis with scattered individually necrotic keratinocytes, (changes identical to those seen in EM) ( Figure 2.17A )

    Figure 2.17, A, Fixed drug eruption. Necrotic keratinocytes in the epidermis, vacuolar alteration of the basal cell layer, and patchy lichenoid inflammatory cell infiltrate that obscures the dermoepidermal junction are seen. Histologic changes are similar to those seen in erythema multiforme. B, Erythema dyschromicum perstans. Interface change with prominent underlying pigment incontinence.

  • Superficial and deep perivascular and lichenoid inflammatory cell infiltrate composed of lymphocytes, eosinophils, and occasional neutrophils

  • Prominent pigment incontinence

  • Bullous variant resulting from more extensive epidermal necrosis has been described

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Erythema Multiforme

    • Interface dermatitis with scattered individually necrotic keratinocytes and a predominantly superficial perivascular and patchy lichenoid lymphoid cell infiltrate

  • Erythema Dyschromicum Perstans/Lichen Planus Pigmentosus

    • Interface dermatitis with scattered individually necrotic keratinocytes; prominent pigment incontinence may be the only feature in older lesions( Figure 2.17B )

Pearls

  • Histopathologic reaction pattern of interface dermatitis not specific or diagnostic and can be exhibited by several unrelated entities

  • Clinicopathologic correlation key to rendering correct diagnosis

  • Fixed drug eruptions occur most commonly with trimethoprim-sulfamethoxazole, acetylsalicylic acid, and phenolphthalein

Selected References

Crowson AN, Magro CM. Recent advances in the pathology of cutaneous drug eruptions. Dermatol Clin . 1999;17:537–560, viii.

Gru AA, Salavaggione AL. Lichenoid and interface dermatoses. Semin Diagn Pathol . 2017;34:237–249.

Sehgal VN, Srivastava G. Fixed drug eruption (FDE): changing scenario of incriminating drugs. Int J Dermatol . 2006;45:897–908.

Lymphomatoid Papulosis

Clinical Features

  • Multiple, small papules that are most often short lived, heal leaving oval scars, and are usually recurrent

Histopathology

  • Wedge-shaped superficial and deep perivascular and lichenoid lymphoid cell infiltrate with admixed eosinophils and scattered large and small atypical lymphocytes

  • Surface ulceration may be present

  • Reed Sternberg–like cells (in type A) and atypical lymphocytes with cerebriform nuclei resembling mycosis fungoides (in type B) are occasionally seen

  • Rarely, a dense monomorphous infiltrate of atypical lymphocytes resembling anaplastic large cell lymphoma (type C) can be seen in association with typical clinical features of lymphomatoid papulosis ( Figure 2.18 )

    Figure 2.18, Lymphomatoid papulosis.

Special Stains and Immunohistochemistry

  • Immunohistochemical stain CD30 highlights the presence of clusters of CD30+ atypical lymphocytes

Other Techniques for Diagnosis

  • Clonal rearrangement of T-cell receptor gene may be present

Differential Diagnosis

  • Insect Bite Reaction

    • Superficial and deep perivascular and interstitial lymphoid cell infiltrate with numerous eosinophils

    • While reactive CD30+ lymphocytes can be seen these are typically single cells and not clustered

  • PLEVA

    • Histologic pattern of interface and lichenoid dermatitis similar to lymphomatoid papulosis

    • Reactive CD30+ lymphocytes can be seen, however, these are typically scattered single cells and not clustered

Pearls

  • Presence of scattered CD30 positive cells not diagnostic of lymphomatoid papulosis as these may be present in reactive conditions

  • Presence of clusters of CD30 positive atypical lymphoid cells and a wedge-shaped inflammatory infiltrate diagnostic for lymphomatoid papulosis

  • Clinical correlates of histopathologic subtypes of lymphomatoid papulosis are of no known relevance

  • Progression of lymphomatoid papulosis to large cell anaplastic lymphoma (CD30 positive) can occur, suggesting that lymphomatoid papulosis may represent the benign end in the spectrum of CD30-positive T-cell lymphoproliferative disorders

Selected References

Cerroni L. Lymphomatoid papulosis, pityriasis lichenoides et varioliformis acuta, and anaplastic large-cell (Ki-1+) lymphoma. J Am Acad Dermatol . 1997;37:287.

Guitart J, Querfeld C. Cutaneous CD30 lymphoproliferative disorders and similar conditions: a clinical and pathologic prospective on a complex issue. Semin Diagn Pathol . 2009;26:131–140.

LeBoit PE. Lymphomatoid papulosis and cutaneous CD30+ lymphoma. Am J Dermatopathol . 1996;18:221–235.

Werner B, Massone C, Kerl H, Cerroni L. Large CD30-positive cells in benign, atypical lymphoid infiltrates of the skin. J Cutan Pathol . 2008;35:1100–1107.

Psoriasiform Dermatitis

Secondary Syphilis

Clinical Features

  • Hematogenous dissemination of causative organism, Treponema pallidum , results in a cutaneous eruption that can be macular, papular, papulosquamous, or, rarely, pustular

  • Associated constitutional symptoms such as fever and lymphadenopathy may be present; other manifestations include condyloma lata, syphilis cornee, lues maligna, and alopecia

Histopathology

  • Patchy/confluent parakeratosis containing neutrophils

  • Regular psoriasiform epidermal hyperplasia (minimal in macular lesions, prominent in condylomata lata) with focal spongiosis ( Figure 2.19A )

    Figure 2.19, Secondary syphilis.

  • Interface change with scattered individually necrotic keratinocytes and papillary dermal edema

  • Superficial and deep perivascular, periadnexal, and lichenoid lymphoplasmacytic infiltrate; plasma cells may be present around nerves ( Figure 2.19B )

  • Infiltrate can be lymphocytic, lymphoplasmacytic, or lymphohistiocytic with rare granuloma formation

Special Stains and Immunohistochemistry

  • Silver stain (Warthin-Starry) may show spirochetes within the epidermis in one third of cases ( not used anymore because of high background associated with this stain )

  • Immunohistochemistry with a monoclonal antibody to T. pallidum yields better detection rates (organisms are more often found in the dermis compared to the epidermis)

Other Techniques for Diagnosis

  • Molecular detection of T. pallidum using PCR the gold standard

Differential Diagnosis

  • Mycosis Fungoides

    • Shows psoriasiform lichenoid pattern similar to syphilis; however, in mycosis fungoides, atypical lymphoid cells are present within the dermal infiltrate and in the mildly spongiotic epidermis

    • Plasma cells are not frequent

  • Subacute and Chronic Spongiotic Dermatitis, Including Photoallergic Dermatitis

    • May show some psoriasiform hyperplasia and spongiosis

    • In general, plasma cells are not prominent

  • Pityriasis Lichenoides

    • Can simulate secondary syphilis but shows predominantly a lymphocytic infiltrate without plasma cells

  • Psoriasis and Psoriasiform Drug Eruption and AIDS-Associated Psoriasiform Dermatitis

    • Inflammatory infiltrate is typically not deep as in secondary syphilis and plasma cells not prominent

    • Suprapapillary plate thinning is not a feature of secondary syphilis

  • Sarcoid and Other Conditions With a Prominent Granulomatous Pattern

    • May appear similar to the granulomatous pattern of secondary syphilis

    • Clinical correlation and serologic studies are necessary

Pearls

  • Increased plasma cells in the inflammatory infiltrate not a constant histopathologic feature

  • The organism is more commonly seen in the dermis and best detected with immunohistochemistry or PCR-based techniques

  • Interface change with a perineural plasma cell-rich infiltrate highly suggestive of secondary syphilis

  • An unusual variant of secondary syphilis is lues maligna, an ulcerative form characterized by thrombotic endarteritis of vessels in the deep dermis resulting in ischemic necrosis

  • Serologic tests are the most common method of diagnosing syphilis

Selected References

Hoang MP, High WA, Molberg KH. Secondary syphilis: a histologic and immunohistochemical evaluation. J Cutan Pathol. 2004;31:595-599.

Jeerapaet P, Ackerman AS. Histologic patterns of secondary syphilis. Arch Dermatol. 1973;107:373.

Muller H, Eisendle K, Brauninger W, et al. Comparative analysis of immunohistochemistry, PCR and focus-floating microscopy for the detection of T. pallidum in mucocutaneous lesions of primary, secondary and tertiary syphilis. Br J Dermatol. 2011;165:50-60.

Nodular And Diffuse Dermatitis

Neutrophils Predominant

Sweet Syndrome

Clinical Features

  • Characterized clinically by fever, arthralgia, leukocytosis, and a cutaneous eruption of painful violaceous plaquelike or nodular lesions involving the extremities and face; truncal lesions are uncommon

  • Affects adults mainly

  • An underlying myeloproliferative disorder, most commonly acute myeloid leukemia or inflammatory disease and rarely solid tumors may be present

  • Sweet syndrome–like eruption reported with many drugs

Histopathology

  • Dense, diffuse, upper dermal predominantly neutrophilic infiltrate, abundant leukocytoclasia, scattered lymphocytes, histiocytes, and eosinophils ( Figure 2.20 )

    Figure 2.20, Sweet syndrome.

  • Marked papillary dermal edema, reactive endothelial cell atypia, and extravasated erythrocytes. Fibrinoid necrosis of vessel walls is not typically seen

  • Histiocytoid variant-infiltrate composed of immature myeloid cells; lesional cells are not neoplastic

Special Stains and Immunohistochemistry

  • Gram stain useful in excluding an infectious etiology such as cellulitis

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Leukocytoclastic Vasculitis

    • Vascular damage with fibrin deposition in the vessel wall seen in leukocytoclastic vasculitis is not a feature of Sweet syndrome

  • Pyoderma Gangrenosum

    • Inflammatory infiltrate (predominately neutrophilic) is deeper and denser than in Sweet syndrome

    • Surface ulceration, foci of folliculitis, and secondary vasculitis may be evident

  • Other Neutrophilic Dermatoses

    • Behçet disease (pathergic lesion), bowel-associated dermatosis-arthritis syndrome, rheumatoid neutrophilic dermatosis—all characterized by a superficial and deep, predominantly neutrophil-rich inflammatory cell infiltrate without vasculitis and minimal papillary dermal edema

Pearls

  • Neutrophil-rich inflammatory infiltrate, leukocytoclasia but NOT vasculitis and marked papillary dermal edema characteristic

  • Histiocytoid variant of Sweet syndrome is not a variant of leukemia cutis

  • Potential infectious etiology should be considered and excluded in all cases of neutrophilic dermatoses

Selected References

  • Buck T., González L.M., Lambert W.C., Schwartz R.A.: Sweet’s syndrome with hematologic disorders: a review and reappraisal. Int J Dermatol 2008; 47: pp. 775-782.
  • Raza S., Kirkland R.S., Patel A.A., et. al.: Insight into Sweet’s syndrome and associated-malignancy: a review of the current literature. Int J Oncol 2013; 42: pp. 1516-1522.
  • Sweet R.D.: Acute febrile neutrophilic dermatosis. Br J Dermatol 1964; 74: pp. 349-356.

Pyoderma Gangrenosum

Clinical Features

  • Idiopathic ulceronecrotic skin disease that begins as follicular papules and pustules that eventually ulcerate

  • Lower extremities and trunk are often involved

  • Fully developed lesions show a necrotic center with a raised, undermined border with a dusky-purple hue

  • Pyoderma gangrenosum may be the cutaneous manifestation of underlying systemic diseases such as inflammatory bowel disease, connective tissue disease, hematopoietic malignancies, and liver diseases

Histopathology

  • Histologic features are nonspecific and not diagnostic—vary according to the age of the lesion and the area biopsied

  • Area biopsied:

    • Center of the lesion shows ulcer with necrosis, dense diffuse neutrophilic infiltrate, and occasionally secondary vasculitis ( Figure 2.21 )

      Figure 2.21, Pyoderma gangrenosum.

    • Undermined border shows a mixed inflammatory cell infiltrate in addition to neutrophils

    • Periphery of the ulcer shows a lymphocytic and histiocytic inflammatory infiltrate

  • Age of the lesion

    • Early lesions show an intradermal microabscess

    • Advanced lesions show a perivascular lymphoplasmacytic infiltrate, reactive endothelial cell atypia and secondary vasculitis

Special Stains and Immunohistochemistry

  • Special stains and microbiologic cultures help exclude an infectious etiology

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Sweet Syndrome

    • Neutrophilic infiltrate that is typically more superficial, leukocytoclasia and marked papillary dermal edema

  • Bacterial Cellulitis

    • Requires demonstration of bacteria by Gram stain or microbiologic cultures

Pearls

  • Histopathology nonspecific and varies with age of the lesion and area biopsied

  • Clinicopathologic correlation important in making the diagnosis and excluding other ulceronecrotic inflammatory disorders of the skin

Selected References

Binus AM, Qureshi AA, Li VW, Winterfield LS. Pyoderma gangrenosum: a retrospective review of patient characteristics, comorbidities and therapy in 103 patients. Br J Dermatol . 2011;165:1244.

Ruocco E, Sangiuliano S, Gravina AG, et al. Pyoderma gangrenosum: an updated review. J Eur Acad Dermatol Venereol . 2009;23:1008.

Su WP, Schroeter AL, Perry HO, et al. Histopathologic and immunopathologic study of pyoderma gangrenosum. J Cutan Pathol . 1983;13:323.

Eosinophils Predominant

Eosinophilic Cellulitis/Wells syndrome

Clinical Features

  • Relatively rare, recurrent dermatosis of uncertain pathogenesis, characterized by sudden onset of erythematous patches that evolve into painful plaques

  • May be idiopathic or associated with insect bites, parasitosis, infections, myeloproliferative disorders, and drug reactions

  • Associated with peripheral eosinophilia in at least 50% of the patients

Histopathology

  • Spongiotic intraepidermal vesicles may be present

  • Dense diffuse dermal infiltrate composed predominantly of eosinophils and occasionally extending into the subcutaneous tissue

  • “Flame figures” (eosinophilic debris caused by degranulating eosinophils and may impregnate the collagen bundles) more prominent in older lesions ( Figure 2.15C )

  • Palisading histiocytes with central necrobiosis may be seen in florid lesions

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Dermal hypersensitivity reactions (to drugs or an arthropod bite)—similar histopathology

Pearls

  • Flame figures although a helpful diagnostic clue not specific for Wells syndrome

  • Eosinophilic cellulitis most likely represents an exaggerated dermal hypersensitivity reaction rather than a specific disease, and a search for inciting stimuli is warranted

Selected References

Brasileiro LG, Abreu MAMM, Paschoal RS. Wells’ syndrome: the importance of differential diagnosis. An Bras Dermatol . 2019;94:370–372.

Caputo R, Marzano AV, Vezzoli P, Lunardon L. Wells syndrome in adults and children: a report of 19 cases. Arch Dermatol . 2006;142:1157.

Fujii K, Tanabe H, Kanno Y, et al. Eosinophilic cellulitis as a cutaneous manifestation of idiopathic hypereosinophilic syndrome. J Am Acad Dermatol . 2003;49:1174–1177.

Scabies

Clinical Features

  • Contagious, pruritic, papulovesicular, and pustular eruption most pronounced on the abdomen, buttocks, and anterior axillary folds caused by the mite Sarcoptes scabiei

  • Eruption believed to be a hypersensitivity reaction

  • Burrows, produced by the female mite, typically involve the palms, web spaces between fingers, and male genitalia

  • Persistent pruritic nodules/nodular scabies involving most commonly the scrotum can be seen up to several months after treatment

Histopathology

  • Sections taken from the burrow show a tunnel-like space (“molting pouches”) between layers of parakeratosis containing the mite/larvae/ova/feces ( Figure 2.22 ).

    Figure 2.22, Scabies.

  • Spongiosis and vesiculation may be present in the epidermis

  • Superficial and deep dermal infiltrate containing varying numbers of eosinophils

  • Persistent nodular lesions show dense, diffuse, mixed inflammatory cell infiltrate containing eosinophils, thick-walled blood vessels, and occasionally atypical mononuclear cells, “pseudolymphomatous” pattern may also be seen. Mite is generally absent in these lesions

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • A suspected burrow can be shaved, placed on a glass slide, and examined under oil immersion

Differential Diagnosis

  • In the absence of the mite or its products in the cornified layer, the histologic changes cannot be distinguished from other hypersensitivity reactions such as those caused by arthropod bites

Pearls

  • Norwegian scabies is a rare variant in which an immeasurable number of mites is present within the cornified layer; generally seen in immunocompromised patients or patients with poor hygiene

  • Scabetic by-product or “feces” in the stratum corneum may be the only clue

  • Pink “pigtails” in the stratum corneum representing egg fragments or casings left behind after the mite hatches may be the only clue

Selected References

Brites C, Weyll M, Pedroso C, et al. Severe and Norwegian scabies are strongly associated with retroviral (HIV-1/HTLV-1) infection in Bahia, Brazil. AIDS . 2002;16:1292.

Chosidow O. Scabies and pediculosis. Lancet . 2000;355:819–826.

Fernandez N, Torres A, Ackerman AB. Pathological findings in human scabies. Arch Dermatol . 1977;113:320.

Histiocytes Predominant

Xanthogranuloma

Clinical Features

  • Benign non-Langerhans cell histiocytosis that most commonly occurs during infancy (within first 6 months of life) but can be seen occasionally in adults (adult xanthogranuloma)

  • About 20% are congenital

  • Usually presents as single or multiple tan to pink-red nodules that almost always regress over time to a tan macule or depression

  • Occasionally found in the deep soft tissue

  • Eye, particularly the uveal tract, most frequent site of extracutaneous involvement

Histopathology

  • Distinctive architecture—exophytic lesion with a well-defined epidermal collarette

  • Lesional cells—uniform-appearing histiocytes with an eosinophilic, vacuolated, or xanthomatous cytoplasm,

  • Touton giant cells are typically seen ( Figure 2.23 )

    Figure 2.23, Xanthogranuloma.

  • Admixture of neutrophils, eosinophils, and lymphocytes

  • Resolving lesion resembles dermatofibroma

Special Stains and Immunohistochemistry

  • Oil red O highlights intracytoplasmic neutral lipids

  • CD68 and factor XIIIa positive

  • CD1a negative; S-100 protein usually negative

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Langerhans Cell Histiocytosis (Eosinophilic Granuloma)

    • Presence of histiocytes positive for CD1a and S-100 protein and admixed eosinophils, electron microscopy demonstrates Birbeck granules

  • Fibrous Histiocytoma (Dermatofibroma)

    • Spindled fibroblasts and histiocytic cells arranged in a storiform pattern

    • Peripheral collagen entrapment and overlying epidermal hyperplasia with pigmentation of basal keratinocytes

  • Xanthoma

    • Composed of sheets of histiocytes containing abundant intracytoplasmic lipid

    • Cholesterol clefts and multinucleate giant cells are typical

Pearls

  • Pathogenesis remains uncertain; believed to be a reactive rather than neoplastic process

  • Not associated with a lipid abnormality

  • Skin lesions almost always regress with time and ultimately appear as a slight depression on the skin surface

  • Overall prognosis is excellent

  • Distinctive architecture with epidermal collarette Touton giant cells not specific for juvenile xanthogranuloma—can also be seen in dermatofibroma and xanthoma

Selected References

Burgdorf WH, Zelger B. JXG, NF1, and JMML: alphabet soup or a clinical issue? Pediatr Dermatol . 2004;21:174.

Dehner LP. Juvenile xanthogranulomas in the first two decades of life: a clinicopathologic study of 174 cases with cutaneous and extracutaneous manifestations. Am J Surg Pathol . 2003;27:579.

Freyer DR, Kennedy R, Bostrom BC, et al. Juvenile xanthogranuloma: forms of systemic disease and their clinical implications. J Pediatr . 1996;129:227–237.

Reticulohistiocytic Granuloma

Clinical Features

  • Typically occurs in adults

  • Most frequently presents as a well-circumscribed red-brown cutaneous nodule with a red-brown to yellow cut surface

  • May present as localized (giant cell reticulohistiocytoma) or systemic disease (multicentric reticulohistiocytosis)

  • Cutaneous reticulohistiocytoma (localized form)—may present as single or multiple skin lesions with clinical features similar to xanthogranuloma

  • Multicentric reticulohistiocytosis (systemic form)—rare, may involve lymph nodes, heart, bone, and joints in addition to widespread skin involvement, may present with progressive erosive arthritis, fever, and weight loss, association with hyperlipidemia, internal malignancies, and autoimmune diseases

Histopathology

  • Essentially similar features in both localized and systemic forms

  • Well-defined infiltrate of multinucleate, uniform epithelioid histiocytes with abundant eosinophilic (“oncocytic”) cytoplasm, multinucleate giant cells with “ground-glass” cytoplasm ( Figure 2.24 )

    Figure 2.24, Reticulohistiocytic granuloma.

  • Infrequent mitotic activity

  • Scattered chronic inflammatory cells

Special Stains and Immunohistochemistry

  • Lesional cells are CD68 positive and S-100 protein and CD1a negative

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Langerhans Cell Histiocytosis

    • Infiltrate of large histiocytoid CD1a and S100 protein positive cells with nuclear grooves

  • Juvenile Xanthogranuloma

    • Distinctive architecture, polymorphous infiltrate, presence of Touton giant cells diagnostic

Pearls

  • Solitary and disseminated form of reticulohistiocytoma exhibit identical histopathology and cytomorphology

  • Predominant population of oncocytic histiocytes and multinucleate giant cells with “ground glass” cytoplasm distinctive

  • Occasional histiocytes with “ground glass” cytomorphology can be seen in other histiocyte rich entities

  • Disseminated reticulohistiocytosis is associated with various malignancies (carcinoma of the breast, colon, or lung) or systemic disease (tuberculosis, diabetes, hypothyroidism)

  • Polyarthritis seen in the disseminated form is due to infiltrate of similar histiocytic cells found in skin around the joints

Selected References

Jung HD, Kim HS, Lee JY, et al. Multicentric reticulohistiocytosis. Acta Derm Venereol . 2013;93:124–125.

Miettinen M, Fetsch JF. Reticulohistiocytoma (solitary epithelioid histiocytoma): a clinicopathologic and immunohistochemical study of 44 cases. Am J Surg Pathol . 2006;30:521.

Snow JL, Muller SA. Malignancy-associated multicentric reticulohistiocytosis: a clinical, histological, and immunophenotypic study. Br J Dermatol . 1995;133:71–76.

Palisading and Necrobiotic Granulomas

Granuloma Annulare

Clinical Features

  • Benign granulomatous process of unknown etiology

  • Occurs most commonly in children and young adults; females more commonly affected than males

  • Predilection for areas of trauma and exposure, typically the dorsal surface of the hands and feet, ankles, knees, and elbows

  • Single or multiple annular dermal plaques with central clearing and raised erythematous borders

  • Spontaneously regress but can recur

Histopathology

  • Diagnostic histopathologic triad—palisaded lymphohistiocytic infiltrate with occasional multinucleate giant cells (some of which can demonstrate elastophagocytosis) minute foci of necrobiosis (mucinous degeneration of the collagen) and increased dermal mucin ( Figure 2.25 )

    Figure 2.25, Granuloma annulare.

  • Typically involves upper and middle dermis, occasionally only the upper or deep dermis

  • Deep variant (subcutaneous granuloma annulare) typically found in a pediatric population

  • Perivascular infiltrates of lymphocytes; eosinophils in varying numbers may be present

  • Occasional neutrophils and nuclear fragmentation in areas of mucinous degeneration

  • Classified as a “blue granuloma” because of the presence of mucinous necrobiosis

Special Stains and Immunohistochemistry

  • Colloidal iron stain highlights mucin

Other Techniques for Diagnosis

  • Noncontributory

Differential Diagnosis

  • Rheumatoid Nodule

    • Characterized by a deeply eosinophilic homogenous foci of fibrinoid degeneration of collagen surrounded by a palisade of lymphohistiocytic infiltrate

    • Typically seen in the deep dermis or subcutis

  • Necrobiosis Lipoidica

    • Biopsy specimens are usually rectangular

    • Foci of basophilic degeneration of collagen stratified between layers of an inflammatory cell infiltrate (“sandwich appearance”) that typically contains plasma cells

    • Involvement of deep dermis is typical

Pearls

  • Diagnostic histopathologic triad (palisaded lymphohistiocytic infiltrate, necrobiosis and increased dermal mucin)

  • Subcutaneous variant of granuloma annulare (pseudorheumatoid nodule) typically presenting in children with deep-seated nodules in the dermis or subcutaneous fat may be difficult to distinguish from rheumatoid nodule

  • A well-known diagnostic pitfall is diagnosing epithelioid sarcoma as granuloma annulare, and vice versa

Selected References

Barren DF, Cootauco MH, Cohen BA. Granuloma annulare: a clinical review. Prim Care Pract . 1997;1:33–39.

Gunes P, Goktay F, Mansur AT, et al. Collagen-elastic tissue changes and vascular involvement in granuloma annulare: a review of 35 cases. J Cutan Pathol . 2009;36:838.

Magro CM, Crowson AN, Regauer S. Granuloma annulare and necrobiosis lipoidica tissue reactions as a manifestation of systemic disease. Hum Pathol . 1996;27:50–56.

Necrobiosis Lipoidica

Clinical Features

  • Degenerative cutaneous disease of unknown etiology

  • More common in women than men

  • Characteristically affects the anterior tibial surface but also has a predilection for the thighs, popliteal areas, and dorsum of the feet and arms

  • Indurated yellow-brown oval plaques with a violaceous border

  • Center of the plaque may later become atrophic with a distinctive yellow waxy hue

  • Very small proportion occur in patients with diabetes mellitus

Histopathology

  • Rectangular contour of biopsy sample, alternating horizontal layers of basophilic degeneration of collagen and an inflammatory infiltrate of histiocytes, lymphocytes, and plasma cells ( Figure 2.26 ) diagnostic (“sandwich” or tiered appearance at scanning magnification)

    Figure 2.26, Necrobiosis lipoidica.

  • Vascular changes (reactive endothelial cell atypia and thickened vessel walls) are features shared with diabetic microangiopathy

  • Zones of dermal sclerosis

  • Sarcoidal type granulomas are seen on occasion

Special Stains and Immunohistochemistry

  • Noncontributory

Other Techniques for Diagnosis

  • DIF may reveal IgM, IgA, C3 and fibrinogen in thickened vessels

Differential Diagnosis

  • Rheumatoid Nodule

    • Deeply eosinophilic homogenous foci of fibrinoid degeneration of collagen surrounded by a palisade of lymphohistiocytic infiltrate typically seen in the deep dermis or subcutis

  • Granuloma Annulare

    • Demarcated zones of necrobiosis with mucin surrounded by a palisade of lymphohistiocytic infiltrate, typically involving the upper half of the dermis

  • Necrobiotic Xanthogranuloma

    • Atrophic epidermis, granulomatous inflammation with Touton giant cells of the deep dermis and subcutis, necrobiosis with cholesterol clefts

Pearls

  • Characteristic tiered appearance at scanning magnification—zones of necrobiosis alternating with layers of an inflammatory infiltrate

  • Presence of plasma cells in the inflammatory infiltrate a helpful clue

  • Less than 1% of the patients with diabetes develop necrobiosis lipoidica

Selected References

Lowitt MH, Dover JS. Necrobiosis lipoidica. J Am Acad Dermatol . 1991;25:735–748.

Magro CM, Crowson AN, Regauer S. Granuloma annulare and necrobiosis lipoidica tissue reactions as a manifestation of systemic disease. Hum Pathol . 1996;27:50–56.

O’Toole EA, Kennedy U, Nolan JJ, et al. Necrobiosis lipoidica: only a minority of patients have diabetes mellitus. Br J Dermatol . 1999;140:283–286.

Rheumatoid Nodule

Clinical Features

  • Chronic deep-seated inflammatory nodule that occurs in patients with rheumatoid arthritis and occasionally in patients with SLE

  • Seen during the course of the disease in 30% to 40% of patients with rheumatoid arthritis

  • Predilection for areas subject to mechanical trauma, typically in para-articular locations, including metacarpophalangeal and proximal interphalangeal joints

  • Solitary or multiple, firm, nontender, freely mobile, large subcutaneous nodules

Histopathology

  • Central areas of homogeneous eosinophilic degeneration of collagen surrounded by a peripheral palisade of histiocytes and lymphocytes ( Figure 2.27 ), located in the subcutaneous tissue and deep dermis

    Figure 2.27, Rheumatoid nodule.

  • Classified as a “red granuloma” because of the central homogenous eosinophilic areas

Special Stains and Immunohistochemistry

  • Noncontributory

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here