Acessibilidade / Reportar erro

Update on vasculitis: an overview and dermatological clues for clinical and histopathological diagnosis - part I How to cite this: Morita TCAB, Trés GFS, Criado RFJ, Sotto MN, Criado PR. Update on vasculitis: an overview and dermatological clues for clinical and histopathological diagnosis - Part I. An Bras Dermatol. 2020;95:352-68. ,☆☆ ☆☆ Study conducted at the Department of Dermatology, Faculdade de Medicina, Universidade de São Paulo and Department of Dermatology, Faculdade de Medicina do ABC, Santo André, SP, Brazil.

Abstract

The term vasculitis refers to the inflammation of vessel walls. It may range in severity from a self-limited disorder in one single organ to a life-threatening disease due to multiple organ failure. It has many causes, although they result in only a few histological patterns of vascular inflammation. Vessels of any type and in any organ can be affected, a fact that results in a broad variety of signs and symptoms. Different vasculitides with indistinguishable clinical presentations have quite different prognosis and treatments. This condition presents many challenges to physicians in terms of classification, diagnosis, appropriate laboratory workup, and treatment. Moreover, it compels a careful follow-up. This article reviews the Chapel-Hill 2012 classification, etiology, recent insights in pathophysiology, some important dermatological clues for the diagnosis and summarizes treatment of some of these complex vasculitis syndromes.

KEYWORDS
Anti-neutrophil cytoplasmic antibody-associated vasculitis; Classification; Diagnosis; Purpura; Systemic vasculitis; Vasculitis; Vasculitis, leukocytoclastic, cutaneous

Introduction

Vasculitides encompass a large group of heterogeneous diseases characterized by inflammatory reaction localized in the vascular wall and perivascular tissues. To the concern of etiology, it is defined as a complex syndrome because several types of vasculitides can be elicited by different stimulus, although most of them are classified as idiopathic. Both, venous or arterial vessels can be affected, in a pattern of single organ or multisystemic disease. Then, dealing with vasculitides often means to study several distinct pathophysiological mechanisms and a wide clinical spectrum. Often several authors rely on the blood vessel caliber to classify them as small, medium or large-vessel vasculitis. However, when it is held alone, this classification fails to recognize diversity within vessels of the same caliber, their specialized roles in several anatomic areas in response to stimuli, injury, and repair that determine disease patterns.11 Sunderkötter CH, Zelger B, Chen KR, Requena L, Piette W, Carlson JA, et al. Nomenclature of cutaneous vasculitis: dermatologic addendum to the 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheumatol. 2018;70:171-84.,22 Hoffman GS, Calabrese LH. Vasculitis: determinants of disease patterns. Nat Rev Rheumatol. 2014;10:454-62.

Cutaneous vasculitides can range in severity from benign, self-limited, short-course skin eruption to life-threatening disease with multiple-organ failure. Otherwise, systemic vasculitis is divided into two main categories: primary vasculitis syndrome and secondary vasculitis syndrome. The first one is caused by unknown etiology inflammation of blood vessels, while the second one is induced by underlying conditions, including connective tissue diseases, cancer, infections, immunization, and drug allergy. In the majority of cases the cutaneous vasculitides syndromes will show a neutrophilic small vessel vasculitis, often called Cutaneous Leukocytoclastic Angiitis (CLA).33 Carlson JA, Chen KR. Cutaneous vasculitis update: small vessel neutrophilic vasculitis syndromes. Am J Dermatopathol. 2006;28:486-506.

The aim of this review is to clarify the recent concepts about the etiology of distinct vasculitides, the classification preconized for the Chapel Hill Consensus Conference (CHCC) and to emphasize the clues on dermatological physical and/or histopathological exam that may help to suggest a specific group of vasculitis in the clinical practice.

Classification of vasculitis

Clinicians have found it convenient to use vessel size as the most important feature to distinguish different forms of vasculitides.22 Hoffman GS, Calabrese LH. Vasculitis: determinants of disease patterns. Nat Rev Rheumatol. 2014;10:454-62. The CHCC published in 2012 proposed the unification of new types of vasculitis syndromes and their associated illness (Table 1).44 Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65:1-11. However, CHCC is only a nomenclature system/nosology and not a classification or a diagnostic system that helps to direct clinical management. Thus, it subdivides vasculitides based on combinations of features that separate different conditions into definable categories.44 Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65:1-11.

Table 1
Classification and definitions of vasculitides adapted from data adopted by the 2012 Chapel Hill Consensus Conference.

Some cutaneous manifestations involving small, medium and/or large vessels can be found not only in the autoimmune diseases, but also in the autoinflammatory diseases. This term was coined to describe an emerging family of conditions characterized by episodes of unprovoked inflammation due to dysregulation of the innate immune system without the primary role of autoreactive T lymphocytes and/or autoantibodies. Skin involvement in autoinflammatory diseases generally resembles urticarial vasculitis or may have features of neutrophilic dermatoses. Additional skin lesions include purpuric exanthema, nonthrombocytopenic purpura, and pyoderma gangrenosum. In some autoinflammatory diseases, it is still unclear whether vasculitis is an integrated clinical manifestation or represents an additional independent disease.55 Carlson JA, Chen KR. Cutaneous vasculitis update: neutrophilic muscular vessel and eosinophilic, granulomatous, and lymphocytic vasculitis syndromes. Am J Dermatopathol. 2007;29:32-43. We have also included these conditions in a proper sub-classification in table 1.

Generally, the size of vessel involvement correlates with clinical morphology on the dermatological exam. Small, predominately superficial vessel involvement results in urticaria, infiltrated erythema, palpable or non-palpable purpura, vesiculobullous and/or pustules lesions. Ulcers, nodules, pitted scars, white atrophy, or livedo racemosa are associated with arterial-muscular vessel involvement, which will be located at dermal-hypodermal interface or within the subcutis. In vasculitides that involve small and medium-size vessels all kind of cutaneous lesions previously described can co-exist in the same patient. Finally, a large number of ulcers, especially when they are extensive and associated with necrosis, which may be consequence of deep-vessel arterial involvement, often predict recurrent vasculitis and systemic disease (Fig. 1).66 Peleg H, Ben-Chetrit E. Vasculitis in the autoinflammatory diseases. Curr Opin Rheumatol. 2017;29:4-11.

Figure 1
Schematic representation of the skin (epidermis, dermis and subcutaneous tissue), types of blood vessels found in upper and lower dermis vascular plexus, vessels diameter, site of main vasculitis syndromes and sufficient depth of biopsies to represent the skin layer in which the disease is localized.

The recognition of pathologic changes in a cutaneous biopsy enables the identification distinct vasculitis conditions and signals the possibility of progression from localized cutaneous to systemic vasculitis.66 Peleg H, Ben-Chetrit E. Vasculitis in the autoinflammatory diseases. Curr Opin Rheumatol. 2017;29:4-11. Skin biopsy is the gold standard method for the diagnosis of cutaneous vasculitis, also allowing differential diagnoses from vasculitis mimics, such as vaso-occlusive conditions and other diseases (Table 2).

Table 2
Classification of vasculitides according to the vessel size and histopathological findings in biopsy specimens.

Clues for vasculitis diagnosis

The clinicians need to keep in mind that cutaneous vasculitis, particularly Cutaneous Small Vessel Vasculitis (CSVV), is often the most common presentation of several vasculitis syndromes.11 Sunderkötter CH, Zelger B, Chen KR, Requena L, Piette W, Carlson JA, et al. Nomenclature of cutaneous vasculitis: dermatologic addendum to the 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheumatol. 2018;70:171-84. It may occur in 3% of Granulomatosis with Polyangiitis (GPA), 13% of Microscopic Polyangiitis (MPA), and 28% of Eosinophilic Granulomatosis with Polyangiitis (EGPA) patients.77 Lane SE, Watts RA, Shepstone L, Scott DG. Primary systemic vasculitis: clinical features and mortality. QJM. 2005;98:97-111. Some steps are essential in the clinical practice for accurate assistance to the patients with vasculitis: (i) meticulous anamnesis and physical exam; (ii) inspection and palpation of the cutaneous and subcutaneous lesions; (iii) a proper cutaneous biopsy is necessary according to the findings of dermatological exam; (iv) accurate propaedeutic and complementary exams must be performed to exclude systemic or internal organ involvement; (v) a work-up including indicated laboratory exams for infectious and parasitic infestations, autoimmune collagen diseases, blood marrow neoplasia and/or organ-solid cancer is useful for the evaluation of its etiology.

An important definition about the nosology of vasculitis in the skin was revised by Sunderkötter et al.11 Sunderkötter CH, Zelger B, Chen KR, Requena L, Piette W, Carlson JA, et al. Nomenclature of cutaneous vasculitis: dermatologic addendum to the 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheumatol. 2018;70:171-84. The authors emphasized that they can be found in distinct clinical settings: (i) a cutaneous component of systemic vasculitides (e.g. cutaneous manifestations of IgA vasculitis), (ii) a skin-limited or skin-dominant expression or variant of a systemic vasculitis (e.g. skin-limited IgA vasculitis), or (iii) a Single-Organ Cutaneous Vasculitis (SOCV) that differs with regard to clinical, laboratory, and pathologic features from recognized systemic vasculitis (e.g. nodular vasculitis). SOCV does not develop full systemic vasculitis, while skin-dominant forms may do it. Then, systemic vasculitides are defined as those one presenting in at least one organ in addition to the skin.

In most cases of CSVV, although nonspecific signs or symptoms of systemic inflammatory reactions, such as leukocytosis, raised C-reactive protein, or arthralgia may occur, significant systemic manifestations are unlikely.11 Sunderkötter CH, Zelger B, Chen KR, Requena L, Piette W, Carlson JA, et al. Nomenclature of cutaneous vasculitis: dermatologic addendum to the 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheumatol. 2018;70:171-84. After a thorough history, review of systems and physical examination, a systematic and targeted laboratory workup should proceed to confirm SOCV. Although no standard protocol for this workup exists, screening tests should aim at elucidating the underlying cause and extent of organ involvement and should be guided by clinical signs and symptoms. Special attention should be paid to any evidence of systemic vasculitis, such as fever, weight loss, and other constitutional symptoms; arthritis; myalgias; abdominal pain, melena, or hematochezia; cough, hemoptysis, or dyspnea; hematuria or frothy urine; sinusitis or rhinitis; and paresthesia, weakness, or foot drop. Clinicians should also ask patients about potential triggers, including preceding infectious symptoms, usage of medications, vaccines and comorbidities.88 Brandt HRC, Criado PR, Arnone M, Sotto MN, Valente NYS. Small vessel cutaneous vasculitis: etiology, pathogenesis, classification and diagnostic criteria - Part I. An Bras Dermatol. 2007;82:387-406.

9 Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44.
-1010 Zanoni G, Girolomoni G, Bonetto C, Trotta F, Häusermann P, Opri R, et al. Single organ cutaneous vasculitis: case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2016;34:6561-71. We summarized in table 3 the red flag symptoms and/or signs, complementary laboratory exams and etiologic factors to workup systemic vasculitis.1111 Muratore F, Pipitone N, Salvarani C, Schmidt WA. Imaging of vasculitis: state of the art. Best Pract Res Clin Rheumatol. 2016;30:688-706.,1212 Bougea A, Anagnostou E, Spandideas N, Triantafyllou N, Kararizou E. An update of neurological manifestations of vasculitides and connective tissue diseases: a literature review. Einstein (Sao Paulo). 2015;13:627-35.

Table 3
Symptoms and signs evaluated to establish the diagnosis of systemic vasculitis and complementary exams needed to identify etiologic factors associated.

Some clues may help to recognize both the size of the vessel involved and the suspect diagnosis. Arterial hypertension may suggest medium-vessel vasculitis of the renal vasculature, as seen in systemic PAN. Cutaneous lesions as palpable purpura (the most frequent type of lesion in CSVV), nonpalpable purpura, pinpoint papules, vesicle, pustules, splinter hemorrhages, and urticaria are often suggestive of small vessel vasculitis. Vesicles and bullous lesions often arise on purpuric macules. A subsequent hemorrhagic pattern is observed, and these lesions may produce ulcerations in the skin. Nonpalpable purpura on the lower extremities can be found in patients with Sjogren syndrome and other conditions that include hypergammaglobulinemic purpura, cryoglobulinemic vasculitis and Waldenström's hypergammaglobulinemic purpura. Urticarial lesions can occur in Systemic Lupus Erythematosus (SLE), Sjogren syndrome, hypocomplementemic urticarial vasculitis syndrome, and EGPA (Fig. 2).1313 Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44.

Figure 2
Different lesions usually seen in patients with small vessel vasculitis. (a) Purpura, petechiae, vesicles and hemorrhagic blisters in the lower limbs in a patient with cryoglobulinemic vasculitis, (b) Urticated sometimes confluent lesions and purpura in the lower limbs very suggestive of urticaria vasculitis.

In medium-sized vessels vasculitides, patients often present with subcutaneous nodules, livedo, large or deep ulcers, papulonecrotic lesions, and digital infarcts as indicative involvement of occlusive or semi-occlusive of medium vessels vasculature (vascular plexus of dermo-hypodermal junction or exclusively of the subcutaneous tissue). Subcutaneous nodules surrounded by a livedoid pattern are more characteristic of Polyarteritis Nodosa (PAN), cutaneous arteritis, sarcoidosis vasculitis, or SLE, rather than Antineutrophil Cytoplasmic Antibodies (ANCA)-Associated Vasculitides (AAV) (Figs. 3 and 4).1313 Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44.

14 Choucair J. Infectious causes of vasculitis in: updates in the diagnosis and treatment of vasculitis. InTech. 2013.

15 Loricera J, Calvo-Río V, Ortiz-Sanjuán F, González-López MA, Fernández-Llaca H, Rueda-Gotor J, et al. The spectrum of paraneoplastic cutaneous vasculitis in a defined population. Medicine (Baltimore). 2013;92:331-43.

16 Tai YJ, Chong AH, Williams RA, Cumming S, Kelly RI. Retrospective analysis of adult patients with cutaneous leukocytoclastic vasculitis. Australas J Dermatol. 2006;47:92-6.

17 Takatu CM, Heringer APR, Aoki V, Valente NYS, de Faria Sanchez PC, de Carvalho JF, et al. Clinicopathologic correlation of 282 leukocytoclastic vasculitis cases in a tertiary hospital: a focus on direct immunofluorescence findings at the blood vessel wall. Immunol Res. 2017;65:395-401.

18 Arora A, Wetter DA, Gonzalez-Santiago TM, Davis MD, Lohse CM. Incidence of leukocytoclasic vasculitis 1996-2010: a population-based study in Olmsted County, Minnesota. Mayo Clin Proc. 2014;89:1515-24.
-1919 Jarrot PA, Kaplanski G. Pathogenesis of ANCA-associated vasculitis: an update. Autoimmun Rev. 2016;15:704-13.

Figure 3
Distinct lesions normally found on the skin of patients with medium vessel vasculitis: (a) Subcutaneous nodules and ulcers in a patients with cutaneous arteritis, (b) Extensive ulcers with areas of necrosis and residual atrophic scars in the lower limbs of a patients with microscopic polyangiitis.

Figure 4
Distinct lesions normally found on the skin of patients with medium vessel vasculitis: (a) Livedo racemosa in the lower limbs, including the dorsum of the feet in a patient with cutaneous arteritis, (b) Digital necrosis in a patients with ANCA-associated vasculitis - granulomatosis with polyangiitis.

Cutaneous lesions of Winkelmann granulomas are characterized by the occurrence of tender, erythematous to violaceous papulonodules, usually symmetrically distributed on the elbows or distal upper extremities and fingers. Rarely, subcutaneous indurated cord-like bands (‘rope sign') have been observed. Histologically, this disease is a variant of CLA with prominent influx of neutrophils with palisading macrophages surrounding a central necrosis composed of basophilic fibrin, degenerative collagen and, of course, neutrophils and neutrophilic debris. However often a true vasculitis is not observed in histopathology sections. This skin reaction pattern was originally described in EGPA disease but subsequently observed in a variety of infectious diseases such as subacute bacterial endocarditis, hepatitis, lymphoproliferative and autoimmune disorders such as GPA, rheumatoid arthritis, inflammatory bowel disease, Takayasu's arteritis and (flaring) SLE.2020 Obermoser G, Sontheimer RD, Zelger B. Overview of common, rare and atypical manifestations of cutaneous lupus erythematosus and histopathological correlates. Lupus. 2010;19:1050-70.

Digital infarcts are commonly seen in vasculitis associated with rheumatoid arthritis but are also seen in PAN and AAV (Fig. 5a).2121 Carlson JA, Chen KR. Cutaneous pseudovasculitis. Am J Dermatopathol. 2007;29:44-55. Retiform purpura in plaques (Fig. 5b) is an uncommon finding of palpable purpura in a livedoid, reticulate or arciform pattern. This arrange is related to the physiological vascular anatomy and results from ischemia-related hemorrhage around a dermal/subcutaneous vessel prior to complete occlusion. Although retiform purpura may occur in a variety of clinical settings, retiform purpura in plaques indicates vascular inflammation and more distinct pathophysiology, as well non-inflammatory conditions, without a true vasculitis, didactically grouped as cutaneous pseudovasculitis (antiphospholipid syndrome, calciphylaxis, warfarin-induced cutaneous necrosis, heparin-induced skin necrosis, cryoglobulinemia, cholesterol or crystal emboli, and oxalosis).2121 Carlson JA, Chen KR. Cutaneous pseudovasculitis. Am J Dermatopathol. 2007;29:44-55.

Figure 5
Retiform purpura in a patient with Propylthiouracil (PTU)-induced antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis: (a) Palpable purpura in a livedoid, reticulate pattern with a necrotic center located in the tighs. The recognition of a retiform purpura pattern indicates that a medium vessel component of vasculitis exists, (b) Large ulcerations with irregular borders, granulating wound bed, areas of necrosis and eschar, and serous discharge.

A general panel of complementary exams suggested by our study group includes several tests, previously cited in table 3 and other, especially for those patients without an obvious cause of vasculitis: complete blood cell count with differential, blood urea nitrogen/creatinine, liver function panel, urinalysis, stool guaiac, serum protein electrophoresis with immunofixation to search for evidence of a paraproteinemia, antistreptolysin-O, HBV/HCV/HIV serologies, cryoglobulins, complement levels (CH50, C3, C4), antinuclear antibody, ANCAs and Rheumatoid Factor (RF).99 Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44.,1313 Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44. If the patient is febrile and has a heart murmur, then blood cultures and echocardiography should be performed. For cases with a medium-sized vessel involvement or signs and symptoms of connective tissue disease, an Antinuclear antibody (ANA) should be obtained.1313 Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44.

C-reactive protein levels usually reflect AAV activity. In both AAV and PAN anti-streptolysin O titers should be evaluated because of the common role of streptococci in these settings. Chest radiology studies, gastrointestinal image exams, gynecological examination for females and urological evaluation form male patients are an adequate screening for malignancy in the absence of any other specific signs or symptoms. High fever, weight loss, bicytopenia or severe anemia, Raynaud's phenomenon (in the absence of connective tissue disease), or cryoglobulins should prompt a more thorough search for malignancy.1313 Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44.

Since only a few vasculitides syndromes have pathognomonic clinical, radiographic and or laboratory findings confident and accurate diagnosis requires a representative skin biopsy to histological confirmation. On the other hand, a biopsy diagnosis of vasculitis cannot stand alone, as it must be correlated with clinical history, physical and laboratory findings and/or angiographic features. For example, a diagnosis of SOCV requires that no evidence of systemic manifestations is found.1010 Zanoni G, Girolomoni G, Bonetto C, Trotta F, Häusermann P, Opri R, et al. Single organ cutaneous vasculitis: case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2016;34:6561-71. If systemic vasculitis is suspected, imaging studies can provide a useful means to determine disease extent and activity, and serology, such as C-reactive protein, erythrocyte sedimentation rate and ANCA levels, and type, can be used to monitor disease activity and predict mortality risk, respectively.

Hence, the classification of cutaneous vasculitis into specific syndromes is better approached morphologically by determining vessel size and main inflammatory response. These histological patterns (Table 2) combined with Direct Immunofluorescent (DIF) examination, ANCA status and findings from work-up for systemic disease, allow for specific diagnosis, and ultimately, a more effective therapy. A neutrophil-predominant small vessel vasculitis primarily affecting upper dermal blood vessels and showing papillary IgA deposits is diagnostic of IgA-vasculitis, whereas a neutrophil-predominant small vessel vasculitis affecting dermal and subcutaneous blood vessels with predominance of IgM vascular deposits would implicate Cryoglobulinemic Vasculitis (CV) or Rheumatoid Vasculitis (RV).2222 Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23.

Clinicians more trained in recognizing elementary cutaneous lesions must be able to perform a representative biopsy of the necessary skin layers. In general, a deep and large punch biopsy (5-6 mm in diameter) or an excision biopsy is the preferred mean to sample vessels of all sizes.2222 Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23. Some key points to a proper cutaneous biopsy are listed below:

  1. a)

    A sample biopsy extending to subcutis taken from the most tender, reddish or purpuric, lesioned skin is the key to obtaining a significant diagnostic result;

  2. b)

    The optimal time for skin biopsy is <48 h after the appearance of a vasculitis lesion. If the biopsy is poorly timed, the pathological features of vasculitis may be absent, a fact that must be considered when interpreting a negative biopsy from a patient whose clinical findings suggest vasculitis;

  3. c)

    Purpuric lesions biopsied in the first 24 h display fibrin deposits within the vessel wall accompanied by neutrophilic infiltration of the wall and surrounding hemorrhage and nuclear debris (leukocytoclasia). After 24 h, neutrophils begin to be replaced by lymphocytes and macrophages. Thus, biopsy of lesions >48 h old, regardless of the underlying form of vasculitis, may show lymphocyte-rich infiltrates, troubling the clinical syndromic vasculitides diagnosis;

  4. d)

    Biopsy specimens should be obtained from non-ulcerated skin lesions due to the frequent finding of incidental vasculitis underlying an ulcer bed. If only superficial ulcers are present, biopsy of the edge is acceptable. In the case of deep, ‘punched out' ulcers, biopsy of the subcutis including the central ulcerated area increases diagnostic yield and recognition of an arterial vasculitis, such as systemic PAN or cutaneous arteritis;

  5. e)

    In case of livedo racemosa, a deep biopsy specimen should be taken from the center of the circular livedo segment because it is where is located the stenosed vessel responsible for the cyanotic periphery. In this setting, level sections are often required to find the focus of vasculitis, which is typically focal and segmental.

Mimouni et at.2323 Mimouni D, Ng PP, Rencic A, Nikolskaia OV, Bernstein BD, Nousari HC. Cutaneous polyarteritis nodosa in patients presenting with atrophie blanche. Br J Dermatol. 2003;148:789-94. reviewed 29 consecutive patients presenting white atrophy, 6 of whom had underlying medium-sized vasculitis consistent with PAN. Three of them had been previously diagnosed as having livedoid vasculopathy on superficial biopsies. So, particularly in the setting of white atrophy repeated and deep biopsies are often necessary to reveal the accurate underlying pathology of necrotizing medium-sized vasculitis in the reticular dermis and the subcutis. Meanwhile, Wohlrab et al.2424 Wohlrab J, Fischer M, Wolter M, Marsch WC. Diagnostic impact and sensitivity of skin biopsies in Sneddon's syndrome. A report of 15 cases. Br J Dermatol. 2001;145:285-8. evaluated the sensitivity of skin biopsies in Sneddon's syndrome. The authors collected 5 deep-punch biopsies (4 mm at least) from different areas where livedo was detected (3 from white and 2 from red areas) in 15 patients. The method had a sensitivity of 27% with one biopsy, 53% with two biopsies and 80% with three biopsies taken from white areas in all cases.

DIF studies should be performed whenever it is possible. For example, the diagnosis of IgA-vasculitis can only be made when IgA vascular deposits are found.2222 Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23. In cases of immune complex-mediated vasculitis, 100% of skin samples will reveal immunoglobulins within the first 48 h, as 70% will do it at 48-72 h. After this interval, despite complement deposits may still be detected in more than 50% of vasculitis skin lesions, immunoglobulins will not.2222 Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23.

Our group performed a retrospective study on the DIF results in a tertiary center in Brazil from January 2007 to December 2014. We evaluated 235 cutaneous samples in 282 patients with CLA. Age ranged from 5 to 87 years-old, with a median age of 45 years. 191/282 (67.73%) of the patients were female. DIF analysis showed positivity in 70.21% of the samples, and C3 was the most frequent immunoreactant. IgA deposition at the blood vessel wall was related to age and absence of autoimmune/inflammatory diseases, while Immunoglobulin M (IgM) deposition was related to female gender, autoimmune/inflammatory disorders, C3 and C4 consumption, and antinuclear antibody or anti-SSA/anti-SSB positivity. Immunoglobulin G (IgG) deposition at the blood vessel wall was associated with age and positive ANCA. Finally, C3 deposition at the blood vessel wall, the most frequent finding of this study, was associated with hematuria and renal involvement. Systemic involvement was present in 12.5% of patients with CLA.1717 Takatu CM, Heringer APR, Aoki V, Valente NYS, de Faria Sanchez PC, de Carvalho JF, et al. Clinicopathologic correlation of 282 leukocytoclastic vasculitis cases in a tertiary hospital: a focus on direct immunofluorescence findings at the blood vessel wall. Immunol Res. 2017;65:395-401.

In summary, in cases of cutaneous vasculitides it is essential to select the proper site of cutaneous biopsy. The histopathological features of CLA may include neutrophils invading the vessel wall, fibrinoid necrosis or fibrin within the vessel wall, nuclear dust (leukocytoclasia), extravasated erythrocytes, perivascular inflammatory cell infiltration, mainly neutrophils, and endothelial swelling. Biopsies for DIF should be taken in the first 8-24 h of appearance of the lesion. Otherwise, the inflammation destroys the immune complexes and they become falsely negative. Detection of immune complex deposition may have diagnostic and prognostic significance. Besides immunoreactants in the vessel wall, DIF can reveal deposition of IgG and C3 at the dermo-epidermal junction, pointing out a positive lupus band test, or may be related to hypocomplementemic urticarial vasculitis with an underlying LES.2222 Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23.

Epidemiology data on vasculitis

The annual incidence of CLA is estimated in 45/million in the United States (population-based in Olmsted County, Minnesota - cutaneous biopsy-proven cases).1818 Arora A, Wetter DA, Gonzalez-Santiago TM, Davis MD, Lohse CM. Incidence of leukocytoclasic vasculitis 1996-2010: a population-based study in Olmsted County, Minnesota. Mayo Clin Proc. 2014;89:1515-24. It affects both genders equally and patients of all ages.99 Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44. In children, by contrast, IgA-vasculitis is much more common than non-IgA small vessel vasculitis of the skin.99 Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44. The presence of an underlying systemic vasculitis, connective tissue disease, or malignancy is much more common in adults than in children.99 Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44.,1818 Arora A, Wetter DA, Gonzalez-Santiago TM, Davis MD, Lohse CM. Incidence of leukocytoclasic vasculitis 1996-2010: a population-based study in Olmsted County, Minnesota. Mayo Clin Proc. 2014;89:1515-24.

In a review on systemic vasculitides, Elefante et al.2525 Elefante E, Bond M, Monti S, Lepri G, Cavallaro E, Felicetti M, et al. One year in review 2018: systemic vasculitis. Clin Exp Rheumatol. 2018;36(Suppl. 111):12-32. presented some data about the epidemiology of AAV: (i) ANCA specificity was associated with different genetic background, clinical features, treatment response, and prognosis in terms of relapse rate and survival rate; (ii) distribution of ANCA specificity differs significantly between the ethnic groups; (iii) MPO-ANCA resulted significantly more frequent in Asian populations [Japanese OR = 59.2 (95% CI 8.0-440.7), p < 0.001, Chinese OR = 6.8 (95% CI 2.6-17.8), p < 0.001], Caucasian American [OR = 2.6 (95% CI 1.7-4.0), p < 0.001] and Middle Eastern/Turkish [OR = 2.3 (95% CI 1.3-4.2), p < 0.005], when compared to the Northern Europeans, who presented more PR3-ANCA specificity; (iv) ANCA negativity was also significantly more frequent in Caucasian Americans than Northern Europeans [OR = 2.0 (95% CI 1.3-3.2), p = 0.002], and (v) there are differences in frequency of organ involvement between the ethnic groups, when compared to Northern Europeans, the Asian population presented significantly less ocular and ear, nose, and throat involvement while renal involvement was significantly less frequent in Caucasian Americans and significantly more frequent in Middle Eastern/Turkish, however interestingly, some differences were not completely determined by the differences in ANCA pattern.

Pathogenesis

We can simplify the mechanisms involved in the pathogenesis of vasculitides into two pathways: (i) immune-complex associated vasculitides and (ii) AAV. However, several factors are involved as: genetic basis, innate and acquired immunity, pathogens, immune tolerance disruption, and “autoantigens”.

The causes of CSVV include infections (20%), inflammatory conditions (15-20%), drug reactions (10-15%), and malignancies (5%).2626 Shavit E, Alavi A, Sibbald RG. Vasculitis - what do we have to know? A review of literature. Int J Low Extrem Wounds. 2018;17:218-26. Disease-inducing or promoting factors are not known for more than half of the cases, and so they are currently classified as idiopathic.1010 Zanoni G, Girolomoni G, Bonetto C, Trotta F, Häusermann P, Opri R, et al. Single organ cutaneous vasculitis: case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2016;34:6561-71. Although non-immunologic factors such as direct infection of endothelial cells can cause vasculitis, most lesions are mediated by immunopathogenic mechanisms. These mechanisms can be classified into Gell and Coombs' four types of hypersensitivity reactions. The majority of cutaneous lesions are likely due to type III hypersensitivity reactions. Immune complexes deposition in postcapillary venules activates complement, which, in turn, induces mast cell degranulation and neutrophil chemotaxis.1010 Zanoni G, Girolomoni G, Bonetto C, Trotta F, Häusermann P, Opri R, et al. Single organ cutaneous vasculitis: case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2016;34:6561-71.

Virtually all drugs have been considered as potential precipitating agents for vasculitis and CSVV is by far the most common form of drug-associated vasculitis. According to the study of Ortiz-Sanjuán et al.2727 Ortiz-Sanjuán F, Blanco R, Hernández JL, Pina T, González-Vela MC, Fernández-Llaca H, et al. Drug-associated cutaneous vasculitis: study of 239 patients from a single referral center. J Rheumatol. 2014;41:2201-7. the most frequently associated were antibiotics, mainly B-lactams and quinolones. Other agents involved were nonsteroidal anti-inflammatory drugs, paracetamol, allopurinol and anticonvulsants. In this series of 239 cases, patients with vasculitis due to major infections, such as endocarditis or pneumonia, were excluded.

RV is probably the most widely recognized form of secondary vasculitis and is typically seen in long standing, erosive, seropositive rheumatoid arthritis. Deposition of immune complexes and subsequent activation of the complement cascade is believed to play a key role in the pathogenesis of RV. An increased expression of inflammatory cytokines such as TNF-α, IL-1 and IL-6 is noted, although their exact role in the pathophysiology of RV remains unclear. Meanwhile, cutaneous vasculitis may be seen in approximately 19-28% of patients with SLE.2828 Sharma A, Dhooria A, Aggarwal A, Rathi M, Chandran V. Connective tissue disorder-associated vasculitis. Curr Rheumatol Rep. 2016;18:31. Small vessels especially post capillary venules are involved in most cases (80-90%).2828 Sharma A, Dhooria A, Aggarwal A, Rathi M, Chandran V. Connective tissue disorder-associated vasculitis. Curr Rheumatol Rep. 2016;18:31. Presence of anti-endothelial antibody is detected in up to 80% of these patients and might contribute by activation of endothelial cells, direct cytotoxic effect due to complement-dependent cytotoxicity or indirect cytotoxic effect secondary to antibody-dependent cytotoxicity.2828 Sharma A, Dhooria A, Aggarwal A, Rathi M, Chandran V. Connective tissue disorder-associated vasculitis. Curr Rheumatol Rep. 2016;18:31.

Genetic susceptibility for vasculitides

Genome-wide association studies have revealed a pivotal role of the Human Leukocyte Antigen (HLA) region in the genetic susceptibility to vasculitides. However, each form has distinct HLA association markers that define them, most likely due to disease-specific differences in antigenic drivers. Despite the considerable advance in the identification of consistent genetic risk factors during the last 10 years, the number of identified risk loci for most types of vasculitides remains significantly lower than other immune-mediated diseases.2929 Carmona FD, Martín J, González-Gay MA. Genetics of vasculitis. Curr Opin Rheumatol. 2015;27:10-7.

There is a strong association with HLA class II alleles with IgA-vasculitis pathogenesis, specifically HLA-DRB1 in Europeans, mainly due to HLA-DR1*0103.43, suggesting that it may be related to other class II vasculitides such as giant cell arteritis or AAV. It has also been proposed that, although less strong, there is a potential effect of HLA class I in the pathogenesis of IgA vasculitis. When it comes to AAV, the genetic distinctions within this group may be related to the antigenic specificity of ANCA rather than to the clinical syndrome. Different AAV subtypes are underpinned by distinct risk factors, with GPA being associated with HLA-DP, SERPINA1, PRTN3 and semaphorin 6A, whereas MPA is mainly associated with HLA-DQ polymorphisms.2929 Carmona FD, Martín J, González-Gay MA. Genetics of vasculitis. Curr Opin Rheumatol. 2015;27:10-7.

30 López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): an updated review. Autoimmun Rev. 2018;17:301-15.
-3131 Lamprecht P, Kerstein A, Klapa S, Schinke S, Karsten CM, Yu X, et al. Pathogenetic and clinical aspects of anti-neutrophil cytoplasmic autoantibody-associated vasculitides. Front Immunol. 2018;9:680.

Takayasu arteritis is predominantly related to HLA class I alleles, in particular to HLA-B52, as it is Behçet's Disease (BD).3030 López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): an updated review. Autoimmun Rev. 2018;17:301-15. The strongest single-risk factor for BD was confirmed to be HLA-B51, but other HLA-B alleles with lower independent effects were also observed, for example, HLA-B15 and HLA-B27.2929 Carmona FD, Martín J, González-Gay MA. Genetics of vasculitis. Curr Opin Rheumatol. 2015;27:10-7. Outside the major histocompatibility complex region, susceptibility to BD has been associated to interleukin 10 and the region of interleukin 23 receptor/interleukin 12 receptor β2.2929 Carmona FD, Martín J, González-Gay MA. Genetics of vasculitis. Curr Opin Rheumatol. 2015;27:10-7.

The association of HLA-DR alleles with PAN has been reported, but with variable results according with the studied population. Unfortunately, there is little information on genetic susceptibility to hypersensitivity vasculitis (CSVV or SCOV). Genetic studies on this vasculitis are scarce and no association of this condition with the HLA region was confirmed, suggesting a heterogeneous etiology in the pathogenesis of this vasculitis that often is restricted and limited to skin.3030 López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): an updated review. Autoimmun Rev. 2018;17:301-15.

Infections as a trigger of vasculitides

Various immune mechanisms are involved in infection-associated vasculitides pathogenesis. Most of them are caused by direct invasion and proliferation of pathogens in the vascular walls causing inflammation. On the other hand, they can sometimes be attributed to indirect immunological effects such as type II, III, or IV hypersensitivity reactions triggered by the infection. Structures such as fimbriae, besides MSCRAMMs (microbial surface components that recognize adhesive matrix molecules) and others cell-wall-anchored proteins are utilized by bacteria to make their way into vulnerable cells. It has been demonstrated that viruses have analogous membrane-adhesion molecules. Nonetheless, whether their density of expression has a role in determining the frequency with which distinct organs are affected is a knowledge to be seek in scientific vasculitides-research related.22 Hoffman GS, Calabrese LH. Vasculitis: determinants of disease patterns. Nat Rev Rheumatol. 2014;10:454-62.,3232 Teng GG, Chatham WW. Vasculitis related to viral and other microbial agents. Best Pract Res Clin Rheumatol. 2015;29:226-43.

Vasculitis has been reported in association with numerous microorganisms, ranging from viruses to fungi. In specific situations, the relationship between etiology and angiocentric inflammation and destruction is clear, as it is in aortitis caused by Mycobacterium tuberculosis or by Treponema pallidum, for which there is a predilection for the ascending aorta. However, other infections, such as HIV, are associated with a wide variety of vasculitic phenotypes, affecting small, medium or large vessels. In these cases, necrotizing, non-necrotizing, giant cell and eosinophilic arteritis have been observed at histology. Direct vessel wall invasion by HIV itself or by opportunistic organisms, and immune complexes containing HIV antigens and antibodies are some of the mechanisms of injure proposed.3333 Haq SA, Pagnoux C. Infection-associated vasculitides. Int J Rheum Dis. 2019;22(Suppl. 1):109-15.

In fact, several infectious diseases are implicated in the production of immune-complex-mediated forms of vasculitis.22 Hoffman GS, Calabrese LH. Vasculitis: determinants of disease patterns. Nat Rev Rheumatol. 2014;10:454-62. IgA vasculitis has been reported to occur in association with various bacteria, including Mycoplasma pneumoniae and Clostridium difficile. As Ureaplasma urealyticum, Aerococcus viridians, Burkholderia cepacia and Listeria monocytogenes have been incriminated in CLA.3333 Haq SA, Pagnoux C. Infection-associated vasculitides. Int J Rheum Dis. 2019;22(Suppl. 1):109-15. Although the incidence of PAN has significantly decreased in the prevailing scenario of the vaccination programs and blood transfusion screening, more than 30% of the cases are secondary to HBV infection. Most evidence supports a type III hypersensitivity injury whereby vascular damage is produced by immune complexes with viral antigens. There have been anecdotal reports of chronic HBV infection associated with other forms of vasculitides such as GPA, even though it still remains unclear whether these reports are causal or coincidental.3232 Teng GG, Chatham WW. Vasculitis related to viral and other microbial agents. Best Pract Res Clin Rheumatol. 2015;29:226-43.

The pathogenesis of HCV-associated vasculitis seems to involve a direct interaction between the virus and lymphocytes leading to polyclonal activation and proliferation of B cells producing IgM with RF activity.3333 Haq SA, Pagnoux C. Infection-associated vasculitides. Int J Rheum Dis. 2019;22(Suppl. 1):109-15. IgM RF is capable of activating the complement system through the binding of the globular domain of the C1qprotein. C1q receptors (gC1q-R) are widely expressed on the surface of blood cells and endothelial cells, and they link to large immunocomplexes containing HCV core protein, facilitating subsequent vascular inflammation. The immunocomplexes with cryoprecipitate containing viral antigens, IgM RF bound to polyclonal anti-HCV IgG, and complement are localized to small vessels of internal organs, although preferentially to the colder extremities.3232 Teng GG, Chatham WW. Vasculitis related to viral and other microbial agents. Best Pract Res Clin Rheumatol. 2015;29:226-43.

HCV-associated CV is a systemic small-to-medium vessel vasculitis due to vascular deposition of cold-precipitable serum proteins, called cryoglobulins. Type I cryoglobulins are monoclonal immunoglobulins, type II cryoglobulins consist of monoclonal immunoglobulins with a RF activity, associated with polyclonal IgG, whereas type III cryoglobulins comprise polyclonal IgG and IgM with RF activity. Elefante et al.2525 Elefante E, Bond M, Monti S, Lepri G, Cavallaro E, Felicetti M, et al. One year in review 2018: systemic vasculitis. Clin Exp Rheumatol. 2018;36(Suppl. 111):12-32. summarized the clinical, laboratorial and predisposing factors influencing the outcome in patients with CV unrelated to HCV: (i) essential (no identifiable underlying disease) CV was the largest group (39.4%) and the first associated condition (21.1%) was primary Sjogren's syndrome (pSS); (ii) overt purpura was present in 78% of patients of pSS group, 64% of whom had type II cryoglobulins, and in patients with pSS the presence of cryoglobulins was associated with highest systemic activity, suggesting that all patients with pSS should be tested for serum cryoglobulins at least at the time of their diagnosis; (iii) SLE-related CV was present in 10.9% of cases as well as other immune conditions, HBsAg positivity in 8.6%, lymphoproliferative disease in 6.8% and some solid tumors in 2.3%; (iv) type II cryoglobulins were present in 54.9% and were independently associated with purpura and fatigue, and (v) older age, male gender, type II cryoglobulins and HBsAg were independently associated with great mortality in patients with HCV-unrelated CV.

PAN is divided into 2 subtypes: systemic and cutaneous. It predominantly presents between 40 and 60 years of age. Besides HBV infection, systemic PAN has been linked to other infectious agents including HCV, HIV, cytomegalovirus, parvovirus B19, and HTLV. The cutaneous PAN has been attributed to streptococcal infections in the pediatric population.2626 Shavit E, Alavi A, Sibbald RG. Vasculitis - what do we have to know? A review of literature. Int J Low Extrem Wounds. 2018;17:218-26.

ANCA-associated vasculitides

The ANCAs are a group of autoantibodies, mainly of the IgG type, developed and released from B cells against antigens in the cytoplasm of neutrophil granulocytes. One type of ANCA is associated with diffuse staining of the cytoplasm and is known as cytoplasmic ANCA (c-ANCA), whilst the other is related with staining around the nucleus and is known as perinuclear ANCA (p-ANCA). The major antigen targeted by c-ANCA is PR3 and that targeted by p-ANCA is MPO. Both are expressed on the cell surface of neutrophils activated by pro-inflammatory cytokines such as IL-1β and TNF generated during infections. ANCAs link to these antigens. Meanwhile, the Crystallizable Fragment (Fc) portion of these ANCAs binds to Fcγ receptors on neutrophils, inducing their excessive activation. This fact leads to abnormal cytokine production, release of Reactive Oxygen Species (ROS) and lytic enzymes, and eventually Neutrophil Extracellular Traps (NET) formation.3434 Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol. 2019;15:91-101.,3535 Suwanchote S, Rachayon M, Rodsaward P, Wongpiyabovorn J, Deekajorndech T, Wright HL, et al. Anti-neutrophil cytoplasmic antibodies and their clinical significance. Clin Rheumatol. 2018;37:875-84.

AAV constitutes a group of rare diseases characterized by necrotizing inflammation of small to medium-sized blood vessels and the presence of ANCA. They are represented by GPA, MPA with its renal limited form pauci-immune necrotizing crescentic glomerulonephritis and EGPA. Results from studies have demonstrated that ANCA, detected by the immunofluorescence technique (c-ANCA or p-ANCA), are a sensitive marker for the so-called AAV, with sensitivity ranging from 80% to more than 90%. Unfortunately, immunofluorescence has a low specificity (80% or less), which is mainly caused by positive p-ANCA in disease controls, such as in inflammatory bowel diseases. P-ANCA, in disease controls, can be also caused by the presence of anti-nuclear antibodies. Antigen specificity (PR3 or MPO) does not effectively differentiate among the AAV, however c-ANCA/PR3-ANCA are mainly found in GPA, while p-ANCA/MPO-ANCA are more prevalent in MPA, pauci-immune necrotizing crescentic glomerulonephritis and CSS. ANCA are detected in 70%-90% of active, generalized GPA, but only in about 40%-50% of the loco-regional forms.3636 Radice A, Bianchi L, Sinico RA. Anti-neutrophil cytoplasmic autoantibodies: methodological aspects and clinical significance in systemic vasculitis. Autoimmun Rev. 2013;12:487-95. Beyond a diagnostic serological marker, data support a pathogenic role for ANCA, which promote activation of primed neutrophils and monocytes, and their adhesion to the endothelium, leading to subsequent tissue damage.1919 Jarrot PA, Kaplanski G. Pathogenesis of ANCA-associated vasculitis: an update. Autoimmun Rev. 2016;15:704-13.

The proteins of complement system participate in the pathogenesis of several small-medium vessels vasculitis including AAV, CV, IgA vasculitis and urticarial vasculitis. PR3- and MPO-ANCA-activated neutrophils prompt C5a release. Subsequent interaction of C5a (a protein derivate from alternative complement pathway activation) with the C5a receptor 1 (C5aR1) may represent a proinflammatory amplification loop in AAV.3030 López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): an updated review. Autoimmun Rev. 2018;17:301-15. Due this complement activation, elevated serum and plasma concentrations of C5a and C3a have been observed in active AAV, especially MPO-ANCA positive vasculitis.3131 Lamprecht P, Kerstein A, Klapa S, Schinke S, Karsten CM, Yu X, et al. Pathogenetic and clinical aspects of anti-neutrophil cytoplasmic autoantibody-associated vasculitides. Front Immunol. 2018;9:680.

Besides microbial components such as peptides or lipopolysaccharides and complement, certain drugs mainly propylthiouracil (PTU), hydralazine and levamisole-adulterated cocaine stimulate ANCA autoantigen expression on the membrane surface of neutrophils. Although these immune cells are the paramount player in the pathogenesis of AAV, since they are both effector cells responsible for endothelial damage, and targets of autoimmunity, it has been demonstrated that some others including CD4 T cells and monocytes are also involved.3535 Suwanchote S, Rachayon M, Rodsaward P, Wongpiyabovorn J, Deekajorndech T, Wright HL, et al. Anti-neutrophil cytoplasmic antibodies and their clinical significance. Clin Rheumatol. 2018;37:875-84.

Neutrophil extracellular traps (NETs) and vasculitis

Neutrophils are able to generate NETs, which are found in a variety of conditions, such as infection, malignancy, atherosclerosis, and as previously mentioned autoimmune diseases, such as AAV. Although several stimuli are responsible for neutrophil activation and NETs formation, a little is known on the exact pathway leading to induction of NET formation.3737 Lee KH, Kronbichler A, Park DD, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16:1160-73. The strongest evidence that NETs actually serve as a source of autoantigens driving autoantibody production in vasculitis comes from studies on drug-induced vasculitis. MPO-ANCA positivity is relatively common in patients treated with propylthiouracil, and some of these patients develop a vasculitis-like syndrome.3838 Söderberg D, Segelmark M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front Immunol. 2016;7:256. MPO and PR3 have been demonstrated within NETs isolated from patients with AAV, and ANCA can further induce NET formation in these patients. ANCA itself also can induce autophagy of neutrophils, which promotes NETosis, a unique form of programmed cell death.3838 Söderberg D, Segelmark M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front Immunol. 2016;7:256.

NETs have been shown to be present not only in skin lesions and thrombi from AAV patients, but also in the circulation. NETs containing pro-inflammatory proteins are thought to contribute to vasculitis by direct damage to endothelial cells, activation of the complement system (especially alternative complement pathway), generating p/c-ANCA, and thrombosis formation.3737 Lee KH, Kronbichler A, Park DD, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16:1160-73. However, Wang et al.3939 Wang H, Sha LL, Ma TT, Zhang LX, Chen M, Zhao MH. Circulating level of neutrophil extracellular traps is not a useful biomarker for assessing disease activity in antineutrophil cytoplasmic antibody-associated vasculitis. PLOS ONE. 2016;11:e0148197. demonstrated that circulating levels of NETs cannot be used as a biomarker to assess disease activity in AAV patients.

Conclusion

A great number of significant contributions have been made on the classification, pathogenesis, clinical sub-setting of cutaneous and systemic vasculitides in this paper. The chief reason for the ongoing research is to strive a personalized medicine based on endotypes rather than phenotypes. The capacity of recognizing integumentary lesions of different vasculitides, allied to other systemic symptoms and signs, an adequate biopsy and proper complementary laboratory and image exams can contribute to the correct diagnosis of this challenging group of diseases.

  • Financial support
    This study was carried out with support from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior - Brasil (CAPES) - Código de Financiamento 001.
  • How to cite this: Morita TCAB, Trés GFS, Criado RFJ, Sotto MN, Criado PR. Update on vasculitis: an overview and dermatological clues for clinical and histopathological diagnosis - Part I. An Bras Dermatol. 2020;95:352-68.
  • ☆☆
    Study conducted at the Department of Dermatology, Faculdade de Medicina, Universidade de São Paulo and Department of Dermatology, Faculdade de Medicina do ABC, Santo André, SP, Brazil.
    Abbreviations:
  • AAV  ANCA-Associated Vasculitis
  • ANCA  Anti-Neutrophil Cytoplasmic Antibodies
  • BD  Behçet's Disease
  • c-ANCA  Cytoplasmic-ANCA
  • CHCC  Chapel Hill Consensus Conference
  • CLA  Leukocytoclastic Angiitis
  • CSVV  Cutaneous Small Vessel Vasculitis
  • CV  Cryoglobulinemic Vasculitis
  • DIF  Direct Immunofluorescent
  • DIV  Drug-Induced Vasculitis
  • EGPA  Eosinophilic Granulomatosis with Polyangiitis
  • GCA  Giant Cell Arteritis
  • GPA  Granulomatosis with Polyangiitis
  • HBV  Hepatitis B Virus
  • HCV  Hepatitis C Virus
  • HLA  Human Leukocyte Antigen
  • IgAV  IgA Vasculitis
  • IgM  Immunoglobulin M
  • IgG  Immunoglobulin G
  • KD  Kawasaki Disease
  • MPA  Microscopic Polyangiitis
  • MPO  Myeloperoxidase
  • NETs  Neutrophil Extracellular Traps
  • PAN  Polyarteritis Nodosa
  • pANCA  Perinuclear-ANCA
  • PR3  Proteinase 3
  • pSS  Primary Sjogren’s Syndrome
  • RF  Rheumatoid Factor
  • RV  Rheumatoid Vasculitis
  • SLE  Systemic Lupus Erythematosus
  • SOCV  Single-Organ Cutaneous Vasculitis
  • TAK  Takayasu Arteritis

References

  • 1
    Sunderkötter CH, Zelger B, Chen KR, Requena L, Piette W, Carlson JA, et al. Nomenclature of cutaneous vasculitis: dermatologic addendum to the 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheumatol. 2018;70:171-84.
  • 2
    Hoffman GS, Calabrese LH. Vasculitis: determinants of disease patterns. Nat Rev Rheumatol. 2014;10:454-62.
  • 3
    Carlson JA, Chen KR. Cutaneous vasculitis update: small vessel neutrophilic vasculitis syndromes. Am J Dermatopathol. 2006;28:486-506.
  • 4
    Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised international Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65:1-11.
  • 5
    Carlson JA, Chen KR. Cutaneous vasculitis update: neutrophilic muscular vessel and eosinophilic, granulomatous, and lymphocytic vasculitis syndromes. Am J Dermatopathol. 2007;29:32-43.
  • 6
    Peleg H, Ben-Chetrit E. Vasculitis in the autoinflammatory diseases. Curr Opin Rheumatol. 2017;29:4-11.
  • 7
    Lane SE, Watts RA, Shepstone L, Scott DG. Primary systemic vasculitis: clinical features and mortality. QJM. 2005;98:97-111.
  • 8
    Brandt HRC, Criado PR, Arnone M, Sotto MN, Valente NYS. Small vessel cutaneous vasculitis: etiology, pathogenesis, classification and diagnostic criteria - Part I. An Bras Dermatol. 2007;82:387-406.
  • 9
    Micheletti RG. Small vessel vasculitis of the skin. In: Systemic vasculitides: current status and perspectives. Cham: Springer International Publishing; 2016. p. 233-44.
  • 10
    Zanoni G, Girolomoni G, Bonetto C, Trotta F, Häusermann P, Opri R, et al. Single organ cutaneous vasculitis: case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine. 2016;34:6561-71.
  • 11
    Muratore F, Pipitone N, Salvarani C, Schmidt WA. Imaging of vasculitis: state of the art. Best Pract Res Clin Rheumatol. 2016;30:688-706.
  • 12
    Bougea A, Anagnostou E, Spandideas N, Triantafyllou N, Kararizou E. An update of neurological manifestations of vasculitides and connective tissue diseases: a literature review. Einstein (Sao Paulo). 2015;13:627-35.
  • 13
    Fiorentino DF. Cutaneous vasculitis. J Am Acad Dermatol. 2003;48:311-44.
  • 14
    Choucair J. Infectious causes of vasculitis in: updates in the diagnosis and treatment of vasculitis. InTech. 2013.
  • 15
    Loricera J, Calvo-Río V, Ortiz-Sanjuán F, González-López MA, Fernández-Llaca H, Rueda-Gotor J, et al. The spectrum of paraneoplastic cutaneous vasculitis in a defined population. Medicine (Baltimore). 2013;92:331-43.
  • 16
    Tai YJ, Chong AH, Williams RA, Cumming S, Kelly RI. Retrospective analysis of adult patients with cutaneous leukocytoclastic vasculitis. Australas J Dermatol. 2006;47:92-6.
  • 17
    Takatu CM, Heringer APR, Aoki V, Valente NYS, de Faria Sanchez PC, de Carvalho JF, et al. Clinicopathologic correlation of 282 leukocytoclastic vasculitis cases in a tertiary hospital: a focus on direct immunofluorescence findings at the blood vessel wall. Immunol Res. 2017;65:395-401.
  • 18
    Arora A, Wetter DA, Gonzalez-Santiago TM, Davis MD, Lohse CM. Incidence of leukocytoclasic vasculitis 1996-2010: a population-based study in Olmsted County, Minnesota. Mayo Clin Proc. 2014;89:1515-24.
  • 19
    Jarrot PA, Kaplanski G. Pathogenesis of ANCA-associated vasculitis: an update. Autoimmun Rev. 2016;15:704-13.
  • 20
    Obermoser G, Sontheimer RD, Zelger B. Overview of common, rare and atypical manifestations of cutaneous lupus erythematosus and histopathological correlates. Lupus. 2010;19:1050-70.
  • 21
    Carlson JA, Chen KR. Cutaneous pseudovasculitis. Am J Dermatopathol. 2007;29:44-55.
  • 22
    Carlson JA. The histological assessment of cutaneous vasculitis. Histopathology. 2010;56:3-23.
  • 23
    Mimouni D, Ng PP, Rencic A, Nikolskaia OV, Bernstein BD, Nousari HC. Cutaneous polyarteritis nodosa in patients presenting with atrophie blanche. Br J Dermatol. 2003;148:789-94.
  • 24
    Wohlrab J, Fischer M, Wolter M, Marsch WC. Diagnostic impact and sensitivity of skin biopsies in Sneddon's syndrome. A report of 15 cases. Br J Dermatol. 2001;145:285-8.
  • 25
    Elefante E, Bond M, Monti S, Lepri G, Cavallaro E, Felicetti M, et al. One year in review 2018: systemic vasculitis. Clin Exp Rheumatol. 2018;36(Suppl. 111):12-32.
  • 26
    Shavit E, Alavi A, Sibbald RG. Vasculitis - what do we have to know? A review of literature. Int J Low Extrem Wounds. 2018;17:218-26.
  • 27
    Ortiz-Sanjuán F, Blanco R, Hernández JL, Pina T, González-Vela MC, Fernández-Llaca H, et al. Drug-associated cutaneous vasculitis: study of 239 patients from a single referral center. J Rheumatol. 2014;41:2201-7.
  • 28
    Sharma A, Dhooria A, Aggarwal A, Rathi M, Chandran V. Connective tissue disorder-associated vasculitis. Curr Rheumatol Rep. 2016;18:31.
  • 29
    Carmona FD, Martín J, González-Gay MA. Genetics of vasculitis. Curr Opin Rheumatol. 2015;27:10-7.
  • 30
    López-Mejías R, Castañeda S, Genre F, Remuzgo-Martínez S, Carmona FD, Llorca J, et al. Genetics of immunoglobulin-A vasculitis (Henoch-Schönlein purpura): an updated review. Autoimmun Rev. 2018;17:301-15.
  • 31
    Lamprecht P, Kerstein A, Klapa S, Schinke S, Karsten CM, Yu X, et al. Pathogenetic and clinical aspects of anti-neutrophil cytoplasmic autoantibody-associated vasculitides. Front Immunol. 2018;9:680.
  • 32
    Teng GG, Chatham WW. Vasculitis related to viral and other microbial agents. Best Pract Res Clin Rheumatol. 2015;29:226-43.
  • 33
    Haq SA, Pagnoux C. Infection-associated vasculitides. Int J Rheum Dis. 2019;22(Suppl. 1):109-15.
  • 34
    Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol. 2019;15:91-101.
  • 35
    Suwanchote S, Rachayon M, Rodsaward P, Wongpiyabovorn J, Deekajorndech T, Wright HL, et al. Anti-neutrophil cytoplasmic antibodies and their clinical significance. Clin Rheumatol. 2018;37:875-84.
  • 36
    Radice A, Bianchi L, Sinico RA. Anti-neutrophil cytoplasmic autoantibodies: methodological aspects and clinical significance in systemic vasculitis. Autoimmun Rev. 2013;12:487-95.
  • 37
    Lee KH, Kronbichler A, Park DD, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev. 2017;16:1160-73.
  • 38
    Söderberg D, Segelmark M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front Immunol. 2016;7:256.
  • 39
    Wang H, Sha LL, Ma TT, Zhang LX, Chen M, Zhao MH. Circulating level of neutrophil extracellular traps is not a useful biomarker for assessing disease activity in antineutrophil cytoplasmic antibody-associated vasculitis. PLOS ONE. 2016;11:e0148197.

Publication Dates

  • Publication in this collection
    24 July 2020
  • Date of issue
    May-Jun 2020

History

  • Received
    30 Mar 2019
  • Accepted
    19 Jan 2020
  • Published
    26 Mar 2020
Sociedade Brasileira de Dermatologia Av. Rio Branco, 39 18. and., 20090-003 Rio de Janeiro RJ, Tel./Fax: +55 21 2253-6747 - Rio de Janeiro - RJ - Brazil
E-mail: revista@sbd.org.br