Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Mar 1.
Published in final edited form as: J Invest Dermatol. 2022 Dec 20;143(3):362–373. doi: 10.1016/j.jid.2022.07.035

HIV-Associated Immune Dysregulation in the Skin: A Crucible for Exaggerated Inflammation and Hypersensitivity

Tafadzwa Chimbetete 1,7, Chloe Buck 1,7, Phuti Choshi 1, Rose Selim 1, Sarah Pedretti 2, Sherrie Jill Divito 3, Elizabeth Jane Phillips 4, Rannakoe Lehloenya 5,6, Jonny Peter 1,2,6
PMCID: PMC9974923  NIHMSID: NIHMS1870188  PMID: 36549954

Abstract

Skin diseases are hallmarks of progressive HIV-related immunosuppression, with severe noninfectious inflammatory and hypersensitivity conditions as common as opportunistic infections. Conditions such as papular pruritic eruption are AIDS defining, whereas delayed immune-mediated adverse reactions, mostly cutaneous, occur up to 100-fold more during HIV infection. The skin, constantly in contact with the external environment, has a complex immunity. A dense, tightly junctioned barrier with basal keratinocytes and epidermal Langerhans cells with antimicrobial, innate-activating, and antigen-presenting functions form the frontline. Resident dermal dendritic, mast, macrophage, and innate lymphoid cells play pivotal roles in directing and polarizing appropriate adaptive immune responses and directing effector immune cell trafficking. Sustained viral replication leads to progressive declines in CD4 T cells, whereas Langerhans and dermal dendritic cells serve as viral reservoirs and points of first viral contact in the mucosa. Cutaneous cytokine responses and diminished lymphoid populations create a crucible for exaggerated inflammation and hypersensitivity. However, beyond histopathological description, these manifestations are poorly characterized. This review details normal skin immunology, changes associated with progressive HIV-related immunosuppression, and the characteristic conditions of immune dysregulation increased with HIV. We highlight the main research gaps and several novel tissue-directed strategies to define mechanisms that will provide targeted approaches to prevention or treatment.

INTRODUCTION

The skin represents an organ of major morbidity during HIV infection, with both infective and inflammatory pathologies common. Certain skin manifestations such as papular pruritic eruption (PPE) or opportunistic skin infections are considered AIDS defining (Garg and Sanke, 2017). Severe cutaneous infections as well as delayed immune-mediated adverse reactions such as Stevens–Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are associated with high morbidity and can be life threatening (Peter et al., 2017). The skin and mucosa, being in constant contact with the external environment, have complex immunity and are also the most common site of HIV viral entry. HIV-related immune pathology, defined most specifically by declining CD4 T cells, progresses with specific changes at cutaneous and mucosal surfaces. This review details normal skin immunology and our current understanding of the cutaneous changes associated with progressive HIV-related infection and immunosuppression and the consequent conditions of immune dysregulation increased among persons living with HIV. We highlight the main research gaps in our pathophysiological characterization of these conditions, particularly delayed immune-mediated adverse reactions, and discuss several novel sites of disease techniques being applied to unpick the specific immune pathways involved.

NORMAL SKIN IMMUNOLOGY

Skin barrier

Skin is composed of three major layers: the epidermis, dermis, and subcutis. Keratinocytes (KCs) are the main cell type in the epidermis. The stratum corneum, which consists of piles of dead KCs (corneocytes) and intercellular lipids, forms the outermost layer of the epidermis and is responsible to the largest extent for barrier function (Kabashima et al., 2019). In the deeper granular layer of the epidermis, physical contact of the KCs is maintained by tight junctions, creating another protective layer that is nearly impermeable to microbes (Coates et al., 2019). However, the presence of skin appendages such as hair follicles and sweat ducts creates a weakness in the armor and a point of entry for low-molecular-weight compounds such as haptens, microorganisms, or small-molecule drugs and chemicals (Kabashima et al., 2019). As such, the skin requires a robust immune system ready to act against pathogens.

Innate defenses

Innate and adaptive immune cell populations residing in normal skin and their protective interactions are highlighted in Figure 1a. KCs have multiple immune functions, expressing pattern recognition receptors, for example, toll-like receptors, which induce secretion of proinflammatory cytokines and chemokines, which further activate skin-resident immune cells or recruit them to the skin (Nestle et al., 2009; Richmond and Harris, 2014). The presence of skin commensals such as Staphylococcus epidermidis also directs KC immune responses and can either induce immune activation or suppress inappropriate immune responses (Lai et al., 2009; Linehan et al., 2018). Mast cells, bearing the receptor MRGPRX2, which is a receptor for host-defense antimicrobial peptides (Chompunud Na Ayudhya et al., 2020), are also important for immune defense. MRGPRX2-mediated mast cell activation helps to clear skin bacterial infection, promotes healing, and protects against reinfection (Chompunud Na Ayudhya et al., 2020). Innate lymphoid cells (ILCs), which include ILC1, ILC2, and ILC3 populations, have recently been identified as an important intermediary between innate and adaptive immune responses. They have many parallel functions to adaptive CD4 T cells and produce T-cell–associated cytokines without stimulation by a specific antigen (Panda and Colonna, 2019; Polese et al., 2020).

Figure 1. A figure showing normal cutaneous immunology and immune dysregulation as a result of HIV infection.

Figure 1.

(a) LCs and KCs, and a small number of CD8+ TRM are immune cells residing in the epidermis, whereas dermal DCs, mast cells, TRM (CD8+/CD4+), macrophages, and ILC populate the dermis. KCs express TLRs that recognize PAMPs and DAMPs and are also a source of AMPs, β-defensins, and cathelicidins. AMPs also activate mast cells through the MRGPRX2 receptors, helping to clear bacterial infections and promote healing. LCs express CD207 (Langerin), which also recognizes PAMPs and also expresses CD1a and MHC class-II molecules, which present antigens to CD4 T cells and cross present to CD8 T cells. In response to pathogens, APCs (LCs or DCs) migrate into skin-draining lymph nodes, presenting antigens to naive CD4 and CD8 T cells and stimulating their clonal expansion and differentiation into Teff cells. Chemokines and cytokines secreted by resident immune cells upregulate the expression of endothelial adhesion molecules (i.e., E-selectin) and skin-homing receptors on other circulating immune cells, which bind specific chemokine ligands (cell receptors 1–4). Teff cells differentiate into memory T cell subsets (TCM, TEM, and TRM). TCM express CCR7 and CD62L, limiting their movement to secondary lymph nodes only. TEM cells lack CCR7 but express tissue-homing addressins, allowing them access into peripheral tissue, that is, skin. TRM cells express markers of tissue residency, that is, CD69 ± CD103; lack CCR7 and CD62L; and reside indefinitely in the skin. (b) HIV infection results in the alteration of both innate and adaptive immune response pathways. LCs and DCs, being the first immune cells to encounter HIV, transmit it to T cells in the skin-draining lymph nodes. CD4+ T cells homing to the skin are depleted, and an expansion of antiviral CD8+ and NK cells follows. A shift toward a Th2 cytokine profile is observed in advanced HIV infection, resulting in increased monocyte and eosinophils infiltration as well as B-cell activation. This results in a state of high immune activation and dysfunction of immunoregulatory mechanisms, leading to the onset of inflammatory and hypersensitivity skin disorders. AMP, antimicrobial peptide; DAMP, damage-associated molecular pattern; DC, dendritic cell; ILC, innate lymphoid cell; KC, keratinocyte; LC, Langerhans cell; MHC, major histocompatibility complex; PAMP, pathogen-associated molecular pattern; TCM, central effector memory T; Teff, effector T; TEM, effector memory T; Th, T helper; TLR, toll-like receptor; TRM, tissue-resident memory.

Skin is home to several antigen-presenting cell (APC) populations, including resident subsets and those recruited during inflammatory responses. The primary role of skin-resident APC populations is to lead off immune responses during antigen encounters through the activation of resident immune cells as well as linking innate with the adaptive immune system (Kupper and Fuhlbrigge, 2004). In the steady state, Langerhans cells (LCs) are the main dendritic cell (DC) subset in the epidermis, whereas CD1a+CD1c+ and CD141hiCD14− DCs constitute dermal DCs (Haniffa et al., 2015). LCs express CD207 (langerin), a C-type lectin receptor that recognizes pathogen-associated molecular patterns, as well as CD1a and major histocompatibility complex class-II molecules, which activate T helper (Th) responses and cross present antigens to CD8 T cells (Klechevsky et al., 2008; Zaba et al., 2009). Similar to LCs, CD1a+CD1c+ DCs are capable of polarizing Th1 and Th2 responses and cross-presenting exogenous antigens to CD8+ T cells (Clausen and Stoitzner, 2015; Haniffa et al., 2015), whereas CD141hiCD14− DCs are superior cross-presenting cells (Haniffa et al., 2012). Macrophages are another population of APCs residing in the skin as CD14+AF-mo-Mac (monocyte) and FXIIIa+CD14+AFhiMac (macrophage) subsets (Haniffa et al., 2015).

Adaptive defenses: resident and circulatory

Activation of T cells in skin-draining lymph nodes results in the production of antigen-specific effector T (Teff) cells. Teff cells migrating to the skin express skin-homing receptors, specifically the carbohydrate epitope cutaneous lymphocyte antigen (CLA), which binds to E-selectin (Kupper and Fuhlbrigge, 2004). These Teff cells further differentiate into memory T cells, which include central memory, effector memory (TEM), and tissue-resident memory (TRM) cells (Sallusto et al., 1999). TRM cells are a memory T-cell subset residing in epithelial barrier tissue, and their role in health and disease is increasingly being appreciated (Park and Kupper, 2015). They lack cell molecules that enable them to migrate into lymph nodes (CCR7 and CD62L) and express markers of tissue residency (CD69 ± CD103) (Schunkert et al., 2021), strategically positioning them for rapid on-site immune protection against known pathogens (Nestle et al., 2009). They are best phenotypically characterized as CD44high, CD3+, CD4+/CD8+, CD45RO+CD69+CLA+CCR7−CD62Llow, and CD103+/− (Schunkert et al., 2021). Similar to T cells, B cells are recruited to the skin through endothelial adhesion molecule and chemokine receptor–ligand interactions (Egbuniwe et al., 2015) and play a role in synthesizing antigen-specific antibodies that are crucial in defense against pathogenic bacterial skin infections such as Staphylococcus aureus.

HIV SKIN IMMUNOLOGY

The main cutaneous immunological changes after HIV infection are highlighted in Figure 1b.

CD4+ T-cell depletion is the hallmark of HIV infection (Okoye and Picker, 2013). In addition, there is a significant decrease in CD4+ T-cell proliferative capacity, increased expression of inhibitory molecules CTLA-4 and PD-1, and an increased percentage of CD4+ T cells undergoing apoptosis (Boasso et al., 2009). An expansion of CD8+ and terminal effector T cells is also reported in HIV-infected normal skin (Galhardo et al., 2004). These cells attempt to control ongoing retroviral infection but also mediate cell/tissue damage, which may contribute to skin disorder onset. For example, CD8+ T-cell–mediated granulysin secretion induces KC cell death as observed in TEN (Chung et al., 2008; Yang et al., 2014). The declining CD4+ T-cell counts associated with HIV infection results in a switch from Th1 to Th2 cytokine polarization, which presents as a progressive decline in the levels of IFN-γ and cytotoxic T lymphocyte functioning and successive incline in IL-4, IL-5, and IgE (Clerici and Shearer, 1994, 1993; Klein et al., 1997).

There is increasing circumstantial evidence that TRM cells play a role in mediating skin disorders, with psoriasis and allergic contact dermatitis being the best described (Cheuk et al., 2014; Clark, 2015; Gaide et al., 2015; Suárez-Fariñas et al., 2011). Within drug reactions, their roles in fixed drug eruptions are best characterized to date (Mizukawa et al., 2002; Schunkert et al., 2021; Teraki and Shiohara, 2003). Despite little evidence in the context of HIV infection, it has been reported in other viral infections such as herpes simplex virus (HSV) that HSV-specific CD8+ cells are retained in the genital mucosa as TRM cells and mediate antiviral function (Zhu et al., 2013, 2007); therefore, it is possible that HIV-specific TRM cells in the skin might be increased during the course of HIV infection and contribute to the onset of skin disorders. It has been proposed that virus-specific TRM cells (i.e., the human herpesvirus family) can cross react with drug-induced self-peptides presented by HLA risk allele, resulting in drug hypersensitivity reactions (Schunkert et al., 2021; White et al., 2015). The function of immunosuppressive regulatory T cells (Tregs) in inflammatory skin lesions is poorly characterized in the context of HIV infection. Outside HIV infection in skin disorders such as psoriasis and TEN, Tregs are reported to be decreased in numbers and suppressor activity (Takahashi et al., 2009; Wölfer et al., 1998). In HIV infection, conflicting data have been reported on the effect of HIV on Treg frequency and suppressive capabilities in circulation (Chevalier and Weiss, 2013).

LCs and dermal DCs are the first cells to encounter HIV at mucosal sites and are therefore the preferred targets for HIV infection (Gray et al., 2020; Miller and Bhardwaj, 2013). Th17 cells that contribute to epithelial barrier integrity are also targets for HIV infection because they have been reported to be depleted in mucosal tissue of the gastrointestinal tract (Brenchley et al., 2008; Klatt and Brenchley, 2010). However, there is limited evidence of cutaneous Th17 cell depletion in HIV infection. Although DCs plays a central role in viral transmission, target cell infection, and presentation of HIV antigens (Manches et al., 2014), other APCs such as macrophages also contribute to increased viral load. Activation of TREM-1 on macrophages by HIV has been shown to prolong macrophage survival, rendering them suitable hosts for HIV latent reservoirs (Campbell et al., 2019; Yuan et al., 2017). Although KCs do not directly become infected with HIV, they are capable of secreting immunomodulatory cytokines that may facilitate viral replication and dissemination (Galhardo et al., 2004). Mast cells also express CCR5 and CXCR4 and have been found to be a source of latently infected HIV (Marone et al., 2016; Sundstrom et al., 2007). Patients with untreated HIV, in particular, have been found to have enhanced non-IgE–mediated responses to fluoroquinolones such as ciprofloxacin, which is a small-molecule ligand for MRGPRX2 (Kelesidis et al., 2010). TAT, a fragment of HIV-1 TAT protein, has also been shown to activate MRGPRX2 (Grimes et al., 2019).

CURRENT STATE OF KNOWLEDGE: SKIN DISORDERS ASSOCIATED WITH HIV

Inflammatory skin disorders

PPE and eosinophilic folliculitis.

PPE often arises during early HIV infection and is therefore used as a marker for early diagnosis of HIV infection (Samanta et al., 2009), and incidence and severity of the disease are reported to be inversely proportional to CD4+ T-cell counts (Uthayakumar et al., 1997). PPE is proposed to be the result of an exaggerated immunological reaction to arthropod bites or stings, with increased total IgE reported in patients who develop the condition (Jiamton et al., 2014). PPE has a clinical overlap with eosinophilic folliculitis (EF) of HIV. EF is thought to be mediated by a Th2 response to unknown pathogen or agents, likely to be Pityrosporum ovale or Demodex folliculorum (follicular mite), an autoimmune reaction to sebocyte or a component of sebum (Brenner et al., 1994; Oladokun et al., 2018b). Elevated levels of IL-4, IL-5, RANTES, and eotaxin have been reported in lesional skin, suggesting a Th2 pattern (Amerio et al., 2001; McCalmont et al., 1995).

Seborrheic dermatitis.

Seborrheic dermatitis (SD) is an inflammatory condition that occurs in up to 40% of HIV-infected patients (Mathes and Douglass, 1985) and only about 3% in HIV-uninfected patients (Fröschl et al., 1990; Mathes and Douglass, 1985; Valia, 2006). In HIV-infected individuals, SD often has a sudden onset, is more severe, and is often recalcitrant to treatment. It occurs early during HIV infection, worsening with decreasing CD4 T-cell counts, and is used as an early marker of HIV infection and disease progression (Ippolito et al., 2000; Uthayakumar et al., 1997). Although the pathogenesis of SD is not well-understood, there is an association with skin colonization with the yeast of the genus Malassezia. As a result of the immune dysregulation in patients with HIV, the immune system is unable to clear the yeast, leading to yeast overgrowth and severe inflammation (Garg and Sanke, 2017). In addition to unhindered Malassezia proliferation, disruption in skin microbiota of the affected SD skin has been reported compared with that in healthy areas, and this disruption is thought to contribute to many other inflammatory skin disorders, including atopic-like dermatitis (AD) (Fercek et al., 2021).

Atopic-like dermatitis (in HIV).

Atopic-like dermatitis is a chronic skin condition characterized by xerosis, pruritus, and inflammation of the skin in genetically susceptible individuals (Oladokun et al., 2018b). It is seen in approximately 30–50% of HIV-infected patients compared with 2–20% in HIV-uninfected individuals (Cedeno-Laurent et al., 2011; Lin and Lazarus, 1995) and more common in children (Sodré et al., 2020). There is no specific stage in HIV disease progression that is associated with the onset of the disease; therefore, it is not used as a diagnostic or prognostic indicator (Garg and Sanke, 2017). It is associated with a Th2 cytokine profile with increased IgE levels, increased eosinophils, and elevated IL-4 and IL-5 cytokine levels (Dlova and Mosam, 2006; Ekpe, 2019; Majors et al., 1997). More recently, patients with AD have been shown to have an NK cell deficiency that appears to get better with treatment (Kabashima and Weidinger, 2020; Mack et al., 2020), and HLA and killer Ig-like receptor genetics may also be important risk factors (Margolis et al., 2021). HIV viremia is associated with functional abnormalities in NK cells and may contribute to the permissive environment for atopic-like dermatitis (Fauci et al., 2005).

Chronic actinic dermatitis.

Chronic actinic dermatitis (CAD) is a rare, persistent, and disfiguring photodermatosis, encompassing a spectrum of skin disorders. HIV infection is associated with higher odds of developing photosensitivity (Bilu et al., 2004; Pappert et al., 1994; Vin-Christian et al., 2000), with CAD, photodistributed drug eruptions, pellagra, and porphyria cutanea tarda the most common spectra of photodermatosis that have been reported (Isaacs et al., 2013; Koch, 2017). The clinical features of CAD, including distribution and morphology, are indistinguishable between HIV-infected and -uninfected persons. HIV-infected men of Fitzpatrick skin types V and VI are predominantly affected (Meola et al., 1997; Mercer et al., 2016; Wong and Khoo, 2005, 2003), and in severe cases, HIV-associated late-stage CAD may present with hypopigmented or vitiligo-like depigmentation (Meola et al., 1997; Mercer et al., 2016). HIV-infected cases usually have significant immunosuppression at presentation (CD4 counts < 200 cells/mm3) (Meola et al., 1997; Wong and Khoo, 2003). The pathogenesis of HIV-associated CAD has not been defined, although CD8+ T cells are thought to play a central role. A decrease in CD4:CD8 ratio in lesional skin has been reported in all forms of CAD (Hamada et al., 2017; Hawk, 2004; Pappert et al., 1994). The antigenic molecules in CAD have been postulated to be DNA, RNA, or molecules related to these (Hawk, 2004; Paek and Lim, 2014).

Psoriasis (in HIV).

Psoriasis is a chronic systemic inflammatory disease with cutaneous manifestations. In HIV-infected individuals, the incidence of psoriasis is higher, it often presents atypically with more exuberant clinical features, and it is often recalcitrant to treatment (Cedeno-Laurent et al., 2011). The severity of the disease correlates with the degree of immunosuppression (Garg and Sanke, 2017; Wölfer et al., 1998). Psoriasis outside of HIV infection is associated with a Th1 cytokine profile (Alpalhão et al., 2019). Given the Th2 switch observed in advanced HIV infection, psoriasis in HIV is considered paradoxical (Alpalhão et al., 2019; Morar et al., 2010), with studies revealing that CD8+ T cells, especially the memory subset, play a role in disease pathogenesis, and thus the Th2 shift might be an oversimplification (Cheuk et al., 2014; Fife et al., 2007; Morar et al., 2010; Smoller et al., 1990; Vissers et al., 2004). An interesting observation is that patients with psoriasis appear to be enriched for genetic variants that protect against HIV-1 disease (Chen et al., 2012). This includes HLA class 1 B-alleles that have been associated with the control of HIV-1 replication as well as heightened expression of HLA–C. In addition, HLA-Bw4-80I and the activating KIR3DS1 have been shown to be associated with long-term nonprogression of HIV as well as increased psoriasis susceptibility (Jiang et al., 2013).

Skin manifestations of immune reconstitution inflammatory syndrome.

Immune reconstitution syndrome is an inflammatory condition to pre-existing microbial, host, or other antigens that may occur when patients with HIV are started on antiretroviral medications (Lehloenya and Meintjes, 2006). Combinational antiretroviral therapy (cART) results in the suppression of HIV viral replication and a decline in the viral load, leading to a recovery in CD4 cell number. The restoration of immunity by cART is beneficial because it reduces opportunistic infections and the need for ongoing treatment. However, immune reconstitution inflammatory syndrome (IRIS) manifests because of this immune restoration, causing temporary worsening of several infections and inflammatory skin disorders (Lawn et al., 2005; Lehloenya and Meintjes, 2006; Oladokun et al., 2018b). IRIS is most common in patients commencing cART with CD4 cell counts <50 cells/mm3. The most common types of skin infections seen as part of IRIS include human papillomavirus, reactivation of varicella-zoster virus, cutaneous mycobacterial infections, or molluscum contagiosum. Inflammatory skin disorders of IRIS include AD and EF (Oladokun et al., 2018c).

Hypersensitivity skin disorders

Maculopapular eruptions.

Maculopapular eruptions (MPEs) or morbilliform rash refers to a rash characterized by flat macules and raised papules on a background of erythema. Causes of MPE include cutaneous adverse reactions and viral infections. MPE is the most common clinical manifestation of cutaneous adverse reactions, although it is usually mild and transient. HIV is a well-recognized cofactor in the elicitation of MPE. MPE has been classified both as a type IVb (Th2) and type IVc (cytotoxic T-cell–mediated) hypersensitivity reaction (Ukoha et al., 2015). Outside HIV infection, immunohistochemistry (IHC) studies have shown that cell infiltrates in MPE are mainly composed of CD4+ and CD8+ T cells, expressing markers of cytotoxic function perforin and granzyme (Yawalkar et al., 2000).

Severe cutaneous adverse reactions.

Severe cutaneous adverse reactions (SCARs) occur at a higher rate in HIV-infected patients than in the general population and cause significant morbidity (Peter et al., 2019). SCARs are type IV hypersensitivity reactions, and in HIV-infected persons, drug reaction with eosinophilia and systemic symptoms (DRESS) and SJS/TEN are the two most frequently encountered treatment-limiting forms (Lehloenya and Dheda, 2012; Peter et al., 2019). SCAR is common in persons living with HIV and occurs at all severities of HIV-associated immunosuppression. Data linking CD4 cell count strata with certain SCAR combinations have been conflicting. For example, early studies identified CD4 cell counts >200 cells/mm3 as a risk factor for nevirapine (NVP)-induced SJS and drug-induced liver injury (Dube et al., 2013; Hasan et al., 2022; Tseng et al., 2014); however, there have been NVP-induced SJS cases with severe immunosuppression (<200 cells/mm3) (Britto and Augustine, 2019) and studies where no association between CD4 counts and disease onset was found (Peters et al., 2010; Phanuphak et al., 2007). Other factors associated with progressive immune dysregulation such as expansion of CD8+ and terminal TEM cells, chronic immune activation associated with excessive levels of inflammatory cytokines, altered Th subset ratios with Th2 skewing, and possible depletion of Treg cells have been proposed/shown to increase the risk of developing SCARs in HIV (Cardone et al., 2018; Peter et al., 2019; Phillips and Mallal, 2018). HLA–gene associations are a risk for developing SCARs and can be population specific owing to allele frequency variations; an example being the HLA-B*58:01 risk allele for allopurinol-induced SJS/TEN and DRESS in Han Chinese, African American, and patients of European ancestry (Fontana et al., 2021; Gonçalo et al., 2013; Goodman and Brett, 2021; Hung et al., 2005; Saito et al., 2016; Zhou et al., 2021).

Abacavir hypersensitivity.

Abacavir is a nucleoside analog reverse transcriptase inhibitor used as part of cART for treating HIV (Borrás-Blasco et al., 2008; Phillips and Mallal, 2007). Abacavir hypersensitivity is a rare but life-threatening reaction, occurring in approximately 3–5% of people receiving treatment (Borrás-Blasco et al., 2008). It is characterized by fever, skin rash, gastrointestinal disorders, and respiratory symptoms, occurring within 6 weeks of abacavir initiation (Hetherington et al., 2001; Phillips and Mallal, 2007). Abacavir hypersensitivity has been shown to be restricted by the HLA-B*57:01 allele (Norcross et al., 2012; Ostrov et al., 2012) and is mediated by CD8+ T-cell activation and subsequent release of inflammatory cytokines. Lesional skin biopsies from both abacavir hypersensitivity reaction and a positive patch test have shown infiltration of CD8+ T cells (Giorgini et al., 2011; Micozzi et al., 2015; Shear et al., 2008). Genetic screening for the HLA-B*57:01 allele for abacavir hypersensitivity is recommended by the U.S. Food and Drug Administration, European Medicines Agency, and Canada Health in routine clinical practice to reduce the risk of developing the reaction (Mallal et al., 2008; Rauch et al., 2006; Wang et al., 2022; Zucman et al., 2007). However, a major burden with HLA screening for several drugs is that although HLA risk allele is necessary, it is not sufficient for the development of drug hypersensitivity because not all patients carrying the risk alleles develop reactions (Peter et al., 2017). Cardone et al. (2018) developed an HLA-B*57:01–transgenic mouse model that showed the role CD4+ T cells play in mediating tolerance to the altered endogenous peptide repertoire induced by abacavir and proposed a mechanism by which CD4+ T cells suppress DC maturation (Cardone et al., 2018; Phillips and Mallal, 2018), thereby potentially explaining why some HLA-B*57:01 carriers tolerate abacavir.

RESEARCH GAPS

Outside of HIV infection, the pathogeneses of the majority of the inflammatory skin disorders outlined earlier are well immunohistologically characterized. In contrast, there is little evidence of the immunological changes at the site of disease in the context of HIV coinfection. Despite existing evidence on the effect of HIV on mucosal surfaces, there is a need to expand further to skin with distinct epithelial barriers, defining both normal and disease-associated microenvironments. A summary of HIV-associated inflammatory and hypersensitivity skin disorders, their proposed mechanism of HIV dysregulation, and current techniques applied to research areas are described in Table 1, with macroscopic images highlighting these characteristic conditions shown in Figure 2ai. The majority of data are limited to histopathology and IHC studies, and there is a lack of advanced cutting-edge sites of disease techniques being applied to understand disease pathogenesis in and outside of HIV infection.

Table 1.

HIV-Associated Inflammatory and Hypersensitivity Skin Disorders, Main Immune Effector Cells and Proposed Mechanisms of HIV Immune Dysregulation, and Current Techniques Applied to the Research Area

Skin Disorder Histological Characteristics Main Immune Effector(s) Proposed Mechanism of HIV Dysregulation Current Techniques Applied to Research Area

Papular pruritic eruptions and eosinophilic folliculitis PPE: perivascular and interstitial infiltrate of lymphocytes and eosinophils
EF: predominant folliculocentric eosinophilic and lymphocytic infiltration
T cells, eosinophils Exaggerated Th2 response to insect bites (does not occur outside of HIV) Limited to IHC studies (Afonso et al., 2012; Amerio et al., 2001; McCalmont et al., 1995; Ramos et al., 2005; Resneck et al., 2004; Rosatelli et al., 2000)
Seborrheic dermatitis Marked hyperkeratosis with confluent parakeratosis, acanthosis, and spongiosis. Yeast forms and exocytosis of lymphocytes also seen T cells, neutrophils Exaggerated Th1 and Th2 response to yeast infection (Malassezia species) Limited to histopathology studies (Cedeno-Laurent et al., 2011; Soeprono et al., 1986)
Atopic-like dermatitis Acanthosis and spongiosis Eosinophils, basophils, mast cells, Th2 cells Exaggerated Th2 cytokine profile, especially increased IL-4, IL-5, and IgE levels Limited to histopathology studies (Cedeno-Laurent et al., 2011; Singh and Rudikoff, 2003)
Chronic actinic dermatitis Spongiosis, variable acanthosis with dermal and perivascular lymphocytes T cells (CD8+) Exaggerated CTL response Limited to IHC studies (Hamada et al., 2017; Meola et al., 1997; Pappert et al., 1994; Sidiropoulos et al., 2014)
Psoriasis Dyskeratotic keratinocytes, irregular acanthosis, perivascular infiltrate of lymphocytes, and macrophages DCs, KCs, and T cells (Th1, Th17, Th22 cells) Exaggerated Th1 cytokine profile (does occur outside of HIV) Limited to IHC (Cedeno-Laurent et al., 2011; Schön and Boehncke, 2005; Smoller et al., 1990) and scRNA-seq studies (outside HIV) (Kim et al., 2021)
Skin manifestations of IRIS Varies with the type of skin infection or inflammatory disorder T cells (Th1, CTL) Exaggerated Th1 responses, and defective Treg activity (number or function) in response to infections or self-antigens Limited to IHC studies (Nelson et al., 2017)
Maculopapular eruptions Spongiosis, mild perivascular lymphocytic infiltrate with eosinophils T cells, LCs, eosinophils Exaggerated Th1 and Th2 responses Limited to histopathology studies (Lapins et al., 1996)
Severe cutaneous adverse reactions DRESS: spongiosis, interface dermatitis, basal vacuolization, lymphocytic & eosinophilic infiltration
SJS/TEN: epidermal necrosis, apoptotic keratinocytes, vacuolar interface dermatitis
DRESS: T cells, eosinophils
SJS/TEN: T cells, NK, and NK T cells
Exaggerated Th1 and Th2 responses Limited to IHC (Yang et al., 2014) and scRNA-seq studies (outside HIV) (Kim et al., 2020)
Abacavir hypersensitivity Perivascular inflammatory infiltrate with eosinophils APCs, T cells (CD8+) Exaggerated Th1 responses Limited to histopathology (Micozzi et al., 2015), IHC (Giorgini et al., 2011), and scRNA-seq studies (Redwood et al., 2019)

Abbreviations: APC, antigen-presenting cell; CTL, cytotoxic T lymphocyte; DC, dendritic cell; DRESS, drug reaction with eosinophilia and systemic symptoms; EF, eosinophilic folliculitis; IHC, immunohistochemistry; IRIS, immune reconstitution inflammatory syndrome; PPE, papular pruritic eruption; scRNA-seq, single-cell RNA sequencing; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis; Th, T helper; Treg, regulatory T.

Figure 2. Montage of images highlighting the inflammatory and hypersensitivity skin disorders associated with HIV.

Figure 2.

(a) PPE presents with an itchy and symmetric papular eruption involving predominantly the extremities and trunk, sparing the palms and soles. Apparent secondary changes include excoriations, ulceration, postinflammatory hyperpigmentation or hypopigmentation, and scarring. (b) EF is characterized by an erythematous papulopustular eruption on the face, neck, upper chest, and back. (c) SD is characterized by dry, flaky, or greasy scales in areas of large and active sebaceous glands such as the scalp, eyebrows, beard area, nasolabial folds, and upper chest. It can progress to result in erythroderma (involving >90% of body surface area) or exfoliative dermatitis. (d) AD presents as erythematous scaly papules and plaques. (e) CAD is characterized by pruritic eczematous or lichenified or even infiltrated patches and plaques that are initially limited to sun-exposed areas, although in severe cases, other areas may be involved and become erythrodermic. (f) Psoriasis is characterized by excessively rapid KC proliferation, and common presentations include plaque, erythrodermic, inverse psoriasis, and reactive arthritis-like psoriasis syndrome. (g) Skin manifestations of IRIS present as a temporary worsening of several infections and inflammatory skin disorders due to the restoration of immunity by cART. (g) The image shows a flare up of molluscum contagiosum after the initiation of cART. (h) MPE is characterized by flat macules and papules on a background of erythema. (i) DRESS (left) has diverse cutaneous lesions, often starting as a symmetrical MPE that may progress to coalescent erythema, infiltrated plaques, purpura, exfoliative dermatitis, and facial edema. The rash is accompanied by fever; internal organ involvement (i.e., hepatitis); and hematologic alterations such as eosinophilia, leukocytosis, and/or lymphocytosis. SJS/TEN (right) is characterized by confluent erythematous macules and atypical target lesions on which blisters occur, leading to skin detachment. This detachment also affects the mucosa and reveals subepithelial erosions that are often hemorrhagic. Images (in a, b, d, f, g, and h) were adapted from the study by Oladokun et al. (2018a). Images (in c, e, and i) were obtained from participants in our IMARI-Africa study on the provision of written informed consent (study approved by the University of Cape Town Research Ethics Committee, reference number R031/2018). The authors affirm that human research participants provided informed consent for the publication of the images in Figure 2. AD, atopic-like dermatitis; CAD, chronic actinic dermatitis; cART, combinational antiretroviral therapy; DRESS, drug reaction with eosinophilia and systemic symptoms; EF, eosinophilic folliculitis; IMARI-Africa, Immune-mediated Adverse Drug Reactions in Africa; IRIS, immune reconstitution inflammatory syndrome; KC, keratinocyte; MPE, maculopapular eruption; PPE, papular pruritic eruption; SD, seborrheic dermatitis; SJS/TEN, Stevens–Johnson syndrome/toxic epidermal necrolysis.

TECHNICAL APPROACHES

Expanded immunohistochemistry and multiplex fluorescent microscopy

Basic immunohistochemistry has been invaluable in the early definition of inflammatory skin disorders, helping to identify the main culprit immune cell populations and some of the molecules they produce/express that may be mediating disease. Expanding immunostaining targets is improving disease characterization and will ensure all possible mediators of disease at the site of disease are uncovered. This includes expanding target immune cell populations (both innate and adaptive) and their associated cytokines, chemokines, and cell surface receptors. Furthermore, defining these in and outside of HIV infection would provide great insight into the role of HIV in disease progression. It must be noted that the identification of certain immune cell populations such as TRM cells remains a challenge owing to their phenotypic heterogeneity and expression of multiple surface markers, precluding the use of standard triple colocalization immunofluorescence approaches (Schunkert et al., 2021). Recent advances in multiplex immunofluorescence microscopy imaging aid in overcoming this barrier (Phillips et al., 2021; Willemsen et al., 2022).

Single-cell immune approaches: move from bulk transcriptomic approaches to single-cell analyses

Single-cell RNA sequencing (scRNA-seq) presents another powerful technique that has been applied to characterize the transcriptome of immune and nonimmune cell populations from lesional and nonlesional skin in inflammatory and hypersensitivity conditions. Transcriptomic changes at the site of disease not only provide insights into the current understanding of disease pathogenic mechanisms but also help to identify key pathogenic T-cell populations, overexpressed genes, and altered cellular pathways; all of which can be translated to clinical implementation of personalized medicine (Shalek and Benson, 2017). In skin lesions of psoriasis, this technique has been used to elucidate gene expression profiles of pathogenic versus regulatory immune cell subsets compared with that in normal skin and define personalized and targeted treatment approaches (Kim et al., 2021). Furthermore, in skin lesions of DRESS, pathway analyses have been applied to identify the Jak–signal transducer and activator of transcription pathways as potential therapeutic targets (Kim et al., 2020). Understanding the transcriptomic profile of normal skin in and outside HIV infection will give more insight into the immune cells and gene expression profiles at the steady state. Further profiling of HIV-infected diseased skin will help to characterize disease-causing cell populations.

Spatial transcriptomics

Despite recent advances in scRNA-seq in identifying isolated cell subpopulations in tissue, the inability to capture the spatial localization of cells within the site of disease tissue has been a major limitation. This spatial information is crucial in understanding the intracellular communication underlying normal and diseased skin (Longo et al., 2021; Rao et al., 2021). Spatial transcriptomics addresses this challenge by physically mapping gene sets expressed in specific cell subsets in the actual tissue, shedding light on the niches enriched for distinct gene sets. This technique is increasingly being utilized in characterizing other skin disorders such as cancer (Ma et al., 2021), but to our knowledge, it has not previously been applied in characterizing inflammatory and hypersensitivity skin disorders outlined in this review. Therefore, integrating the scRNA-seq approach together with a physically mapped-out transcriptomic atlas of normal skin in and outside of HIV will further supplement the nature and local distribution of transcriptomic changes observed in diseased skin.

CONCLUSIONS AND FUTURE DIRECTIONS

It is well-established that HIV infection increases the risk of developing varying inflammatory skin disorders. Several HIV-driven immunological changes have been proposed and described systemically and at the level of the skin. The homeostatic function of both immune (innate and adaptive) and nonimmune cell subsets residing in the skin is altered during HIV infection, and this local immune dysregulation is the main driver for the onset of inflammatory skin disorders. The direct mechanistic pathway of HIV at the site of disease in developing specific skin disorders remains elusive, and there are limited research approaches being applied at the site of disease to investigate this. Future research efforts need to focus on applying these more advanced single-cell and spatial transcriptomic approaches, in addition to expanded immunohistochemistry and multiplex approaches, to better understand the specific changes in the skin immunity that drive inflammatory and hypersensitivity disorders, with the potential to develop novel biomarkers and intervention strategies.

ACKNOWLEDGMENTS

The Immune-mediated Adverse Drug Reactions-Africa project is part of the European and Developing Countries Clinical Trials Partnership 2 program supported by the European Union (grant number TMA2017SF-1981). The Immune-mediated Adverse Drug Reactions-South Africa Registry and Biorepository is supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (NIH) under award number R01AI152183. JGP is supported by an NIH Fogarty career development award (K43TW011178-04). TC receives financial support from the Departmental Research Committee of the University of Cape Town Faculty of Health Sciences and an MSc fellowship funded by the NIH (5 D43 TW010559). CB and RS receive financial support from the European and Developing Countries Clinical Trials Partnership. PC is supported by the NIH Fogarty PhD fellowship (5 D43 TW010559) and the South African Medical Research Council through its Division of Research Capacity Development under the Bongani Mayosi National Health Scholars Programme. RL’s work is supported by the South African Medical Research Council and nonrated researcher support from the South African National Research Foundation. EJP reports grants from the NIH (P50GM115305, R01HG010863, R01AI152183, U01AI154659, R13AR078623, UAI109565) and the National Health and Medical Research Council of Australia. She receives Royalties from Uptodate and consulting fees from Janssen, Vertex, Biocryst, and Regeneron. She is codirector of IIID Pty that holds a patent for HLA-B*57:01 testing for abacavir hypersensitivity and has a patent pending for Detection of Human Leukocyte Antigen-A*32:01 in connection with Diagnosing Drug Reaction with Eosinophilia and Systemic Symptoms without any financial remuneration and not directly related to the submitted work. We acknowledge Karen Adamson, a freelance graphics designer, from Cape Town, South Africa, who helped with figure illustrations.

Abbreviations:

AD

atopic dermatitis

APC

antigen-presenting cell

CAD

chronic actinic dermatitis

cART

combinational antiretroviral therapy

CLA

cutaneous lymphocyte antigen

DC

dendritic cell

DRESS

drug reaction with eosinophilia and systemic symptoms

EF

eosinophilic folliculitis

HSV

herpes simplex virus

IHC

immunohistochemistry

ILC

innate lymphoid cell

IRIS

immune reconstitution inflammatory syndrome

KC

keratinocyte

LC

Langerhans cell

MPE

maculopapular eruption

NVP

nevirapine

PPE

papular pruritic eruption

SCAR

severe cutaneous adverse reaction

scRNA

single-cell RNA sequencing

SD

seborrheic dermatitis

SJS

Stevens–Johnson syndrome

Teff

effector T cell

TEM

effector memory T

TEN

toxic epidermal necrolysis

Th

T helper

Treg

regulatory T cell

TRM

tissueresident memory

Footnotes

CONFLICT OF INTEREST

SJD is/has been a consultant for MEI Pharma, Adaptimmune, and Sanofi; none are related to the enclosed work. The remaining authors state no conflict of interest.

DISCLAIMER

Funders played no role in any aspect of this review.

REFERENCES

  1. Afonso JP, Tomimori J, Michalany NS, Nonogaki S, Porro AM. Pruritic papular eruption and eosinophilic folliculitis associated with human immunodeficiency virus (HIV) infection: a histopathological and immunohistochemical comparative study. J Am Acad Dermatol 2012;67:269–75. [DOI] [PubMed] [Google Scholar]
  2. Alpalhão M, Borges-Costa J, Filipe P. Psoriasis in HIV infection: an update. Int J STD AIDS 2019;30:596–604. [DOI] [PubMed] [Google Scholar]
  3. Amerio P, Verdolini R, Proietto G, Feliciani C, Toto P, Shivji G, et al. Role of Th2 cytokines, RANTES and eotaxin in AIDS-associated eosinophilic folliculitis. Acta Derm Venereol 2001;81:92–5. [DOI] [PubMed] [Google Scholar]
  4. Bilu D, Mamelak AJ, Nguyen RH, Queiroz PC, Kowalski J, Morison WL, et al. Clinical and epidemiologic characterization of photosensitivity in HIV-positive individuals. Photodermatol Photoimmunol Photomed 2004;20:175–83. [DOI] [PubMed] [Google Scholar]
  5. Boasso A, Shearer GM, Chougnet C. Immune dysregulation in human immunodeficiency virus infection: know it, fix it, prevent it? J Intern Med 2009;265:78–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Borrás-Blasco J, Navarro-Ruiz A, Borrás C, Casterá E. Adverse cutaneous reactions associated with the newest antiretroviral drugs in patients with human immunodeficiency virus infection. J Antimicrob Chemother 2008;62:879–88. [DOI] [PubMed] [Google Scholar]
  7. Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, Asher TE, et al. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood 2008;112:2826–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Brenner S, Wolf R, Ophir J. Eosinophilic pustular folliculitis: a sterile folliculitis of unknown cause? J Am Acad Dermatol 1994;31:210–2. [DOI] [PubMed] [Google Scholar]
  9. Britto GR, Augustine M. Mucocutaneous manifestations of human immunodeficiency virus (HIV) infection in children in relation to the degree of immunosuppression. Int J Dermatol 2019;58:1165–71. [DOI] [PubMed] [Google Scholar]
  10. Campbell GR, To RK, Spector SA. TREM-1 protects HIV-1-infected macrophages from apoptosis through maintenance of mitochondrial function. mBio 2019;10. e02638–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cardone M, Garcia K, Tilahun ME, Boyd LF, Gebreyohannes S, Yano M, et al. A transgenic mouse model for HLA-B*57:01-linked abacavir drug tolerance and reactivity. J Clin Invest 2018;128:2819–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Cedeno-Laurent F, Gómez-Flores M, Mendez N, Ancer-Rodríguez J, Bryant JL, Gaspari AA, et al. New insights into HIV-1-primary skin disorders. J Int AIDS Soc 2011;14:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Chen H, Hayashi G, Lai OY, Dilthey A, Kuebler PJ, Wong TV, et al. Psoriasis patients are enriched for genetic variants that protect against HIV-1 disease. PLoS Genet 2012;8:e1002514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cheuk S, Wikén M, Blomqvist L, Nylén S, Talme T, Ståhle M, et al. Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. J Immunol 2014;192:3111–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Chevalier MF, Weiss L. The split personality of regulatory T cells in HIV infection. Blood 2013;121:29–37. [DOI] [PubMed] [Google Scholar]
  16. Chompunud Na Ayudhya C, Roy S, Thapaliya M, Ali H. Roles of a mast cell-specific receptor MRGPRX2 in Host Defense and Inflammation. J Dent Res 2020;99:882–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chung WH, Hung SI, Yang JY, Su SC, Huang SP, Wei CY, et al. Granulysin is a key mediator for disseminated keratinocyte death in Stevens-Johnson syndrome and toxic epidermal necrolysis. Nat Med 2008;14:1343–50. [DOI] [PubMed] [Google Scholar]
  18. Clark RA. Resident memory T cells in human health and disease. Sci Transl Med 2015;7:269rv1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Clausen B, Stoitzner P. Functional specialization of the skin dendritic cell subsets in regulating T cell responses. Front Immunol 2015;6:534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Clerici M, Shearer GM. A TH1–>TH2 switch is a critical step in the etiology of HIV infection. Immunol Today 1993;14:107–11. [DOI] [PubMed] [Google Scholar]
  21. Clerici M, Shearer GM. The Th1-Th2 hypothesis of HIV infection: new insights. Immunol Today 1994;15:575–81. [DOI] [PubMed] [Google Scholar]
  22. Coates M, Lee MJ, Norton D, MacLeod AS. The skin and intestinal microbiota and their specific innate immune systems. Front Immunol 2019;10:2950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Dlova NC, Mosam A. Inflammatory noninfectious dermatoses of HIV. Dermatol Clin 2006;24:439e48. vi. [DOI] [PubMed] [Google Scholar]
  24. Dube N, Adewusi E, Summers R. Risk of nevirapine-associated StevensJohnson syndrome among HIV-infected pregnant women: the Medunsa National Pharmacovigilance Centre, 2007e2012. S Afr Med J 2013;103:322–5. [DOI] [PubMed] [Google Scholar]
  25. Egbuniwe IU, Karagiannis SN, Nestle FO, Lacy KE. Revisiting the role of B cells in skin immune surveillance. Trends Immunol 2015;36:102–11. [DOI] [PubMed] [Google Scholar]
  26. Ekpe O Pruritic papular eruption of HIV: a review article. Our Dermatol Online 2019;10:191–6. [Google Scholar]
  27. Fauci AS, Mavilio D, Kottilil S. NK cells in HIV infection: paradigm for protection or targets for ambush [published correction appears in Nat Rev Immunol 2005;5:960] Nat Rev Immunol 2005;5:835–43. [DOI] [PubMed] [Google Scholar]
  28. Fercek I, Lugovic-Mihic L, Tambic-Andrasevic A, Cesic D, Grginic AG, Beslic I, et al. Features skin microbiota common inflam skin diseases. Life (Basel) 2021;11:962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Fife DJ, Waller JM, Jeffes EW, Koo JY. Unraveling the paradoxes of HIV-associated psoriasis: a review of T-cell subsets and cytokine profiles. Dermatol Online J 2007;13:4. [PubMed] [Google Scholar]
  30. Fontana RJ, Li YJ, Phillips E, Saeed N, Barnhart H, Kleiner D, et al. Allopurinol hepatotoxicity is associated with human leukocyte antigen class I alleles. Liver Int 2021;41:1884–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Fröschl M, Land HG, Landthaler M. Seborrheic dermatitis and atopic eczema in human immunodeficiency virus infection. Semin Dermatol 1990;9:230–2. [PubMed] [Google Scholar]
  32. Gaide O, Emerson RO, Jiang X, Gulati N, Nizza S, Desmarais C, et al. Common clonal origin of central and resident memory T cells following skin immunization. Nat Med 2015;21:647–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Galhardo MC, Alvarenga FF, Schueler G, Perez M, Morgado MG, Ferreira H, et al. Normal skin of HIV-infected individuals contains increased numbers of dermal CD8 T cells and normal numbers of Langerhans cells. Braz J Med Biol Res 2004;37:745–53. [DOI] [PubMed] [Google Scholar]
  34. Garg T, Sanke S. Inflammatory dermatoses in human immunodeficiency virus. Indian J Sex Transm Dis AIDS 2017;38:113–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Giorgini S, Martinelli C, Tognetti L, Carocci A, Giuntini R, Mastronardi V, et al. Use of patch testing for the diagnosis of abacavir-related hypersensitivity reaction in HIV patients. Dermatol Ther 2011;24:591–4. [DOI] [PubMed] [Google Scholar]
  36. Gonçalo M, Coutinho I, Teixeira V, Gameiro AR, Brites MM, Nunes R, et al. HLA-B*58:01 is a risk factor for allopurinol-induced DRESS and StevensJohnson syndrome/toxic epidermal necrolysis in a Portuguese population. Br J Dermatol 2013;169:660–5. [DOI] [PubMed] [Google Scholar]
  37. Goodman CW, Brett AS. Race and pharmacogenomics-personalized medicine or misguided practice? JAMA 2021;325:625–6. [DOI] [PubMed] [Google Scholar]
  38. Gray CM, O’Hagan KL, Lorenzo-Redondo R, Olivier AJ, Amu S, Chigorimbo-Murefu N, et al. Impact of chemokine C-C ligand 27, foreskin anatomy and sexually transmitted infections on HIV-1 target cell availability in adolescent South African males. Mucosal Immunol 2020;13:118–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Grimes J, Desai S, Charter NW, Lodge J, Moita Santos RM, Isidro-Llobet A, et al. MrgX2 is a promiscuous receptor for basic peptides causing mast cell pseudo-allergic and anaphylactoid reactions. Pharmacol Res Perspect 2019;7:e00547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hamada T, Aoyama Y, Shirafuji Y, Iwatsuki K. Phenotypic analysis of circulating T-cell subset and its association with burden of skin disease in patients with chronic actinic dermatitis: a hematologic and clinicopathologic study of 20 subjects. Int J Dermatol 2017;56:540–6. [DOI] [PubMed] [Google Scholar]
  41. Haniffa M, Gunawan M, Jardine L. Human skin dendritic cells in health and disease. J Dermatol Sci 2015;77:85–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Haniffa M, Shin A, Bigley V, McGovern N, Teo P, See P, et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity 2012;37:60–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Hasan M, Yunihastuti E, Teguh HK, Abdullah M. Incidence and predictors of nevirapine and efavirenz-associated rash among Indonesian HIV patients. Asian Pac J Allergy Immunol 2022;40:141–6. [DOI] [PubMed] [Google Scholar]
  44. Hawk JL. Chronic actinic dermatitis. Photodermatol Photoimmunol Photomed 2004;20:312–4. [DOI] [PubMed] [Google Scholar]
  45. Hetherington S, McGuirk S, Powell G, Cutrell A, Naderer O, Spreen B, et al. Hypersensitivity reactions during therapy with the nucleoside reverse transcriptase inhibitor abacavir. Clin Ther 2001;23:1603–14. [DOI] [PubMed] [Google Scholar]
  46. Hung SI, Chung WH, Liou LB, Chu CC, Lin M, Huang HP, et al. HLA-b*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol [published correction appears in Proc Natl Acad Sci USA 2005;102:6237] Proc Natl Acad Sci USA 2005;102:4134–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Ippolito F, Passi S, Di Carlo A. Is seborrhoeic dermatitis a clinical marker of HIV disease? Minerva Ginecol 2000;52:54–8. [PubMed] [Google Scholar]
  48. Isaacs T, Ngwanya MR, Dlamini S, Lehloenya RJ. Annular erythema and photosensitivity as manifestations of efavirenz-induced cutaneous reactions: a review of five consecutive cases. J Antimicrob Chemother 2013;68:2871–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Jiamton S, Kaewarpai T, Ekapo P, Kulthanan K, Hunnangkul S, Boitano JJ. Total IgE, mosquito saliva specific IgE and CD4+ count in HIV-infected patients with and without pruritic papular eruptions. Asian Pac J Allergy Immunol 2014;32:53–9. [DOI] [PubMed] [Google Scholar]
  50. Jiang Y, Chen O, Cui C, Zhao B, Han X, Zhang Z, et al. KIR3DS1/L1 and HLA-Bw4–80I are associated with HIV disease progression among HIV typical progressors and long-term nonprogressors. BMC Infect Dis 2013;13:405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Kabashima K, Honda T, Ginhoux F, Egawa G. The immunological anatomy of the skin. Nat Rev Immunol 2019;19:19–30. [DOI] [PubMed] [Google Scholar]
  52. Kabashima K, Weidinger S. NK cells as a possible new player in atopic dermatitis. J Allergy Clin Immunol 2020;146:276–7. [DOI] [PubMed] [Google Scholar]
  53. Kelesidis T, Fleisher J, Tsiodras S. Anaphylactoid reaction considered ciprofloxacin related: a case report and literature review. Clin Ther 2010;32:515–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kim D, Kobayashi T, Voisin B, Jo JH, Sakamoto K, Jin SP, et al. Targeted therapy guided by single-cell transcriptomic analysis in drug-induced hypersensitivity syndrome: a case report. Nat Med 2020;26:236–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Kim J, Lee J, Kim HJ, Kameyama N, Nazarian R, Der E, et al. Single-cell transcriptomics applied to emigrating cells from psoriasis elucidate pathogenic versus regulatory immune cell subsets. J Allergy Clin Immunol 2021;148:1281–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Klatt NR, Brenchley JM. Th17 cell dynamics in HIV infection. Curr Opin HIV AIDS 2010;5:135–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, Thompson-Snipes L, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity 2008;29:497–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Klein SA, Dobmeyer JM, Dobmeyer TS, Pape M, Ottmann OG, Helm EB, et al. Demonstration of the Th1 to Th2 cytokine shift during the course of HIV-1 infection using cytoplasmic cytokine detection on single cell level by flow cytometry. AIDS 1997;11:1111–8. [DOI] [PubMed] [Google Scholar]
  59. Koch K Photosensitive disorders in HIV. South Afr J HIV Med 2017;18:676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kupper TS, Fuhlbrigge RC. Immune surveilance in the skin: mechanisms and clinical consequences. Nat Rev Immunol 2004;4:211–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, Yang Y, Cogen AL, et al. Commensal bacteria regulate toll-like receptor 3–dependent inflammation after skin injury. Nat Med 2009;15:1377–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Lapins J, Lindbäck S, Lidbrink P, Biberfeld P, Emtestam L, Gaines H. Mucocutaneous manifestations in 22 consecutive cases of primary HIV-1 infection. Br J Dermatol 1996;134:257–61. [PubMed] [Google Scholar]
  63. Lawn SD, Bekker LG, Miller RF. Immune reconstitution disease associated with mycobacterial infections in HIV-infected individuals receiving antiretrovirals. Lancet Infect Dis 2005;5:361–73. [DOI] [PubMed] [Google Scholar]
  64. Lehloenya R, Meintjes G. Dermatologic manifestations of the immune reconstitution inflammatory syndrome. Dermatol Clin 2006;24:549–70. [DOI] [PubMed] [Google Scholar]
  65. Lehloenya RJ, Dheda K. Cutaneous adverse drug reactions to anti-tuberculosis drugs: state of the art and into the future. Expert Rev Anti Infect Ther 2012;10:475–86. [DOI] [PubMed] [Google Scholar]
  66. Lin RY, Lazarus TS. Asthma and related atopic disorders in outpatients attending an urban HIV clinic. Ann Allergy Asthma Immunol 1995;74:510–5. [PubMed] [Google Scholar]
  67. Linehan JL, Harrison OJ, Han SJ, Byrd AL, Vujkovic-Cvijin I, Villarino AV, et al. Non-classical immunity controls microbiota impact on skin immunity and tissue repair. Cell 2018;172:784e96–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet 2021;22:627–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ma F, Hughes TK, Teles RMB, Andrade PR, de Andrade Silva BJ, Plazyo O, et al. The cellular architecture of the antimicrobial response network in human leprosy granulomas. Nat Immunol 2021;22:839–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Mack MR, Brestoff JR, Berrien-Elliott MM, Trier AM, Yang TB, McCullen M, et al. Blood natural killer cell deficiency reveals an immunotherapy strategy for atopic dermatitis. Sci Transl Med 2020:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Majors MJ, Berger TG, Blauvelt A, Smith KJ, Turner ML, Cruz PD Jr. HIV-related eosinophilic folliculitis: a panel discussion. Semin Cutan Med Surg 1997;16:219–23. [DOI] [PubMed] [Google Scholar]
  72. Mallal S, Phillips E, Carosi G, Molina JM, Workman C, Tomazic J, et al. HLA-b*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008;358:568–79. [DOI] [PubMed] [Google Scholar]
  73. Manches O, Frleta D, Bhardwaj N. Dendritic cells in progression and pathology of HIV infection. Trends Immunol 2014;35:114–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Margolis DJ, Mitra N, Hoffstad OJ, Kim BS, Monos DS, Phillips EJ. Association of KIR genes and MHC Class I ligands with atopic dermatitis. J Immunol 2021;207:1522–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Marone G, Varricchi G, Loffredo S, Galdiero MR, Rivellese F, de Paulis A. Are basophils and mast cells masters in HIV infection? Int Arch Allergy Immunol 2016;171:158–65. [DOI] [PubMed] [Google Scholar]
  76. Mathes BM, Douglass MC. Seborrheic dermatitis in patients with acquired immunodeficiency syndrome. J Am Acad Dermatol 1985;13:947–51. [DOI] [PubMed] [Google Scholar]
  77. McCalmont TH, Altemus D, Maurer T, Berger TG. Eosinophilic folliculitis. The histologic spectrum. Am J Dermatopathol 1995;17:439–46. [DOI] [PubMed] [Google Scholar]
  78. Meola T, Sanchez M, Lim HW, Buchness MR, Soter NA. Chronic actinic dermatitis associated with human immunodeficiency virus infection. Br J Dermatol 1997;137:431–6. [PubMed] [Google Scholar]
  79. Mercer JJ, Yin NC, Lee ES, Mosam A, Dlova NC. Photodermatitis with subsequent vitiligo-like leukoderma in HIV infection. Int J Dermatol 2016;55:e306–7. [DOI] [PubMed] [Google Scholar]
  80. Micozzi S, Rojas P, Rodriguez-Gamboa A, De Barrio M. Exuberant positive patch test to abacavir in a patient with the HLA-b*5701 haplotype. J Allergy Clin Immunol Pract 2015;3:965–7. [DOI] [PubMed] [Google Scholar]
  81. Miller E, Bhardwaj N. Dendritic cell dysregulation during HIV-1 infection. Immunol Rev 2013;254:170–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Mizukawa Y, Yamazaki Y, Teraki Y, Hayakawa J, Hayakawa K, Nuriya H, et al. Direct evidence for interferon-gamma production by effector-memory-type intraepidermal T cells residing at an effector site of immunopathology in fixed drug eruption. Am J Pathol 2002;161:1337–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Morar N, Willis-Owen SA, Maurer T, Bunker CB. HIV-associated psoriasis: pathogenesis, clinical features, and management. Lancet Infect Dis 2010;10:470–8. [DOI] [PubMed] [Google Scholar]
  84. Nelson AM, Manabe YC, Lucas SB. Immune reconstitution inflammatory syndrome (IRIS): what pathologists should know. Semin Diagn Pathol 2017;34:340–51. [DOI] [PubMed] [Google Scholar]
  85. Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ. Skin immune sentinels in health and disease. Nat Rev Immunol 2009;9:679–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Norcross MA, Luo S, Lu L, Boyne MT, Gomarteli M, Rennels AD, et al. Abacavir induces loading of novel self-peptides into HLA-b*57: 01: an autoimmune model for HLA-associated drug hypersensitivity. AIDS 2012;26:F21–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Okoye AA, Picker LJ. CD4(+) T-cell depletion in HIV infection: mechanisms of immunological failure. Immunol Rev 2013;254:54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Oladokun RE, Lehloenya RJ, Hlela C, Ubesie AC, Katibi SO, Malande OO, et al. Atlas of paediatric HIV infection. Cape Town, South Africa: University of Cape Town libraries; 2018a. [Google Scholar]
  89. Oladokun RE, Lehloenya RJ, Hlela C, Ubesie AC, Katibi SO, Malande OO, et al. Inflammatory skin conditions. In: Oladokun RE, Lehloenya RJ, editors. Atlas of paediatric HIV infection. Cape Town, South Africa: University of Cape Town libraries; 2018b. p. 83–109. [Google Scholar]
  90. Oladokun RE, Lehloenya RJ, Hlela C, Ubesie AC, Katibi SO, Malande OO, et al. Skin manifestations of immune reconstitution inflammatory syndrome (IRIS). In: Oladokun RE, Lehloenya RJ, editors. Atlas of paediatric HIV infection. Cape Town, South Africa: University of Cape Town libraries; 2018c. p. 127. [Google Scholar]
  91. Ostrov DA, Grant BJ, Pompeu YA, Sidney J, Harndahl M, Southwood S, et al. Drug hypersensitivity caused by alteration of the MHC-presented self-peptide repertoire. Proc Natl Acad Sci USA 2012;109:9959–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Paek SY, Lim HW. Chronic actinic dermatitis. Dermatol Clin 2014;32:355–61. [DOI] [PubMed] [Google Scholar]
  93. Panda S, Colonna M. Innate Lympid cells in mucosal immunity. Front Immunol 2019;10:861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Pappert A, Grossman M, DeLeo V. Photosensitivity as the presenting illness in four patients with human immunodeficiency viral infection. Arch Dermatol 1994;130:618–23. [PubMed] [Google Scholar]
  95. Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med 2015;21:688–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Peter J, Choshi P, Lehloenya RJ. Drug hypersensitivity in HIV infection. Curr Opin Allergy Clin Immunol 2019;19:272–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Peter JG, Lehloenya R, Dlamini S, Risma K, White KD, Konvinse KC, et al. Severe delayed cutaneous and systemic reactions to drugs: A global perspective on the science and art of current practice. J Allergy Clin Immunol Pract 2017;5:547–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Peters PJ, Stringer J, McConnell MS, Kiarie J, Ratanasuwan W, Intalapaporn P, et al. Nevirapine-associated hepatotoxicity was not predicted by CD4 count ≥250 cells/μL among women in Zambia, Thailand and Kenya. HIV Med 2010;11:650–60. [DOI] [PubMed] [Google Scholar]
  99. Phanuphak N, Apornpong T, Teeratakulpisarn S, Chaithongwongwatthana S, Taweepolcharoen C, Mangclaviraj S, et al. Nevirapine-associated toxicity in HIV-infected Thai men and women, including pregnant women. HIV Med 2007;8:357–66. [DOI] [PubMed] [Google Scholar]
  100. Phillips D, Schürch CM, Khodadoust MS, Kim YH, Nolan GP, Jiang S. Highly multiplexed phenotyping of immunoregulatory proteins in the tumor microenvironment by CODEX tissue imaging. Front Immunol 2021;12:687673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Phillips E, Mallal S. Drug hypersensitivity in HIV. Curr Opin Allergy Clin Immunol 2007;7:324–30. [DOI] [PubMed] [Google Scholar]
  102. Phillips EJ, Mallal SA. Active suppression rather than ignorance: tolerance to abacavir-induced HLA-B*57:01 peptide repertoire alteration. J Clin Invest 2018;128:2746–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Polese B, Zhang H, Thurairajah B, King I. Innate lymphocytes in psoriasis. Fronitiers Immunol 2020;11:242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Ramos H, Pagliari C, Takakura CF, Sotto MN, Duarte MI. Pruritic papular eruption associated with HIV-etiopathogenesis evaluated by clinical, immunohistochemical, and ultrastructural analysis. J Dermatol 2005;32:549–56. [DOI] [PubMed] [Google Scholar]
  105. Rao A, Barkley D, França GS, Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature 2021;596:211–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Rauch A, Nolan D, Martin A, McKinnon E, Almeida C, Mallal S. Prospective genetic screening decreases the incidence of abacavir hypersensitivity reactions in the Western Australian HIV cohort study. Clin Infect Dis 2006;43:99–102. [DOI] [PubMed] [Google Scholar]
  107. Redwood AJ, Rwandamuriye F, Chopra A, Leary S, Ram R, McDonnell W, et al. Single-cell transcriptomics reveal polyclonal memory T-cell responses in skin with positive abacavir patch test results. J Allergy Clin Immunol 2019;144:1413–6.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Resneck JS Jr, Van Beek M, Furmanski L, Oyugi J, LeBoit PE, Katabira E, et al. Etiology of pruritic papular eruption with HIV infection in Uganda. JAMA 2004;292:2614–21. [DOI] [PubMed] [Google Scholar]
  109. Richmond JM, Harris JE. Immunology and skin in health and disease. Cold Spring Harb Perspect Med 2014;4. a015339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Rosatelli JB, Soares FA, Roselino AM. Pruritic papular eruption of the acquired immunodeficiency syndrome: predominance of CD8+ cells. Int J Dermatol 2000;39:873–4. [DOI] [PubMed] [Google Scholar]
  111. Saito Y, Stamp LK, Caudle KE, Hershfield MS, McDonagh EM, Callaghan JT, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for human leukocyte antigen B (HLA-B) genotype and allopurinol dosing: 2015 update. Clin Pharmacol Ther 2016;99:36–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 1999;401:708–12. [DOI] [PubMed] [Google Scholar]
  113. Samanta M, Kundu C, Sarkar M, Bhattacharyya S, Chatterjee S. Papular pruritic eruptions: A marker of progressive HIV disease in children: experience from eastern India. Indian J Sex Transm Dis AIDS 2009;30:79–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Schön MP, Boehncke WH. Psoriasis. N Engl J Med 2005;352:1899–912. [DOI] [PubMed] [Google Scholar]
  115. Schunkert EM, Shah PN, Divito SJ. Skin resident memory T cells may play critical role in delayed-type drug hypersensitivity reactions. Front Immunol 2021;12:654190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Shalek AK, Benson M. Single-cell analyses to tailor treatments. Sci Transl Med 2017;9. eaan4730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Shear NH, Milpied B, Bruynzeel DP, Phillips EJ. A review of drug patch testing and implications for HIV clinicians. AIDS 2008;22:999–1007. [DOI] [PubMed] [Google Scholar]
  118. Sidiropoulos M, Deonizio J, Martinez-Escala ME, Gerami P, Guitart J. Chronic actinic dermatitis/actinic reticuloid: a clinicopathologic and immunohistochemical analysis of 37 cases. Am J Dermatopathol 2014;36:875–81. [DOI] [PubMed] [Google Scholar]
  119. Singh F, Rudikoff D. HIV-associated pruritus: etiology and management. Am J Clin Dermatol 2003;4:177–88. [DOI] [PubMed] [Google Scholar]
  120. Smoller BR, McNutt NS, Gray MH, Krueger J, Hsu A, Gottlieb AB. Detection of the interferon-gamma-induced protein 10 in psoriasiform dermatitis of acquired immunodeficiency syndrome. Arch Dermatol 1990;126:1457–61. [PubMed] [Google Scholar]
  121. Sodré CS, Rodrigues PMG, Vieira MS, Marques Paes da Silva A, Gonçalves LS, Ribeiro MG, et al. Oral mycobiome identification in atopic dermatitis, leukemia, and HIV patients - a systematic review. J Oral Microbiol 2020;12:1807179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Soeprono FF, Schinella RA, Cockerell CJ, Comite SL. Seborrheic-like dermatitis of acquired immunodeficiency syndrome. a clinicopathologic study. J Am Acad Dermatol 1986;14:242–8. [DOI] [PubMed] [Google Scholar]
  123. Suárez-Fariñas M, Fuentes-Duculan J, Lowes MA, Krueger JG. Resolved psoriasis lesions retain expression of a subset of disease-related genes. J Invest Dermatol 2011;131:391–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Sundstrom JB, Ellis JE, Hair GA, Kirshenbaum AS, Metcalfe DD, Yi H, et al. Human tissue mast cells are an inducible reservoir of persistent HIV infection. Blood 2007;109:5293–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Takahashi R, Kano Y, Yamazaki Y, Kimishima M, Mizukawa Y, Shiohara T. Defective regulatory T cells in patients with severe drug eruptions: timing of the dysfunction is associated with the pathological phenotype and outcome. J Immunol 2009;182:8071–9. [DOI] [PubMed] [Google Scholar]
  126. Teraki Y, Shiohara T. IFN-gamma-producing effector CD8+ T cells and IL-10-producing regulatory CD4+ T cells in fixed drug eruption. J Allergy Clin Immunol 2003;112:609–15. [DOI] [PubMed] [Google Scholar]
  127. Tseng YT, Yang CJ, Chang SY, Lin SW, Tsai MS, Liu WC, et al. Incidence and risk factors of skin rashes and hepatotoxicity in HIV-infected patients receiving nevirapine-containing combination antiretroviral therapy in Taiwan. Int J Infect Dis 2014;29:12–7. [DOI] [PubMed] [Google Scholar]
  128. Ukoha U, Pandya A, Dominguez A. Mobilliform drug eruptions. Cutaneous drug eruptions. London: Springer; 2015. p. 45–53. [Google Scholar]
  129. Uthayakumar S, Nandwani R, Drinkwater T, Nayagam AT, Darley CR. The prevalence of skin disease in HIV infection and its relationship to the degree of immunosuppression. Br J Dermatol 1997;137:595–8. [DOI] [PubMed] [Google Scholar]
  130. Valia RG. Etiopathogenesis of seborrheic dermatitis. Indian J Dermatol Venereol Leprol 2006;72:253–5. [DOI] [PubMed] [Google Scholar]
  131. Vin-Christian K, Epstein JH, Maurer TA, McCalmont TH, Berger TG. Photosensitivity in HIV-infected individuals. J Dermatol 2000;27:361–9. [DOI] [PubMed] [Google Scholar]
  132. Vissers WH, Arndtz CH, Muys L, Van Erp PE, de Jong EM, van de Kerkhof PC. Memory effector (CD45RO+) and cytotoxic (CD8+) T cells appear early in the margin zone of spreading psoriatic lesions in contrast to cells expressing natural killer receptors, which appear late. Br J Dermatol 2004;150:852–9. [DOI] [PubMed] [Google Scholar]
  133. Wang CW, Preclaro IAC, Lin WH, Chung WH. An updated review of genetic associations with severe adverse drug reactions: translation and implementation of pharmacogenomic testing in clinical practice. Front Pharmacol 2022;13:886377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. White KD, Chung WH, Hung SI, Mallal S, Phillips EJ. Evolving models of the immunopathogenesis of T cell-mediated drug allergy: the role of host, pathogens, and drug response. J Allergy Clin Immunol 2015;136:219–34. quiz 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Willemsen M, Tio D, Krebbers G, Kasiem FR, Jaspars EH, Matos TR, et al. Presence of skin tissue-resident memory T cells in human nonmalignant and premalignant melanocytic skin lesions and in melanoma. Am J Dermatopathol 2022;44:416–23. [DOI] [PubMed] [Google Scholar]
  136. Wölfer LU, Djemadji-Oudjiel N, Hiletework M, Tebbe B, Husak R, Goerdt S, et al. [HIV-associated psoriasis. Clinical and histological observations in 36 patients]. Hautarzt 1998;49:197–202. [DOI] [PubMed] [Google Scholar]
  137. Wong SN, Khoo LSW. Chronic actinic dermatitis as the presenting feature of HIV infection in three Chinese males. Clin Exp Dermatol 2003;28:265–8. [DOI] [PubMed] [Google Scholar]
  138. Wong SN, Khoo LSW. Analysis of photodermatoses seen in a predominantly Asian population at a photodermatology clinic in Singapore. Photodermatol Photoimmunol Photomed 2005;21:40–4. [DOI] [PubMed] [Google Scholar]
  139. Yang C, Mosam A, Mankahla A, Dlova N, Saavedra A. HIV infection predisposes skin to toxic epidermal necrolysis via depletion of skin-directed CD4+ T cells. J Am Acad Dermatol 2014;70:1096–102. [DOI] [PubMed] [Google Scholar]
  140. Yawalkar N, Egli F, Hari Y, Nievergelt H, Braathen LR, Pichler WJ. Infiltration of cytotoxic T cells in drug-induced cutaneous eruptions. Clin Exp Allergy 2000;30:847–55. [DOI] [PubMed] [Google Scholar]
  141. Yuan Z, Fan X, Staitieh B, Bedi C, Spearman P, Guidot DM, et al. HIV-related proteins prolong macrophage survival through induction of Triggering receptor expressed on myeloid cells-1. Sci Rep 2017;7:42028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Zaba LC, Krueger JG, Lowes MA. Resident and “inflammatory” dendritic cells in human skin. J Invest Dermatol 2009;129:302–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Zhou Y, Krebs K, Milani L, Lauschke VM. Global frequencies of clinically important HLA alleles and their implications for the cost-effectiveness of preemptive pharmacogenetic testing. Clin Pharmacol Ther 2021;109:160–74. [DOI] [PubMed] [Google Scholar]
  144. Zhu J, Koelle DM, Cao J, Vazquez J, Huang ML, Hladik F, et al. Virus-specific CD8+ T cells accumulate near sensory nerve endings in genital skin during subclinical HSV-2 reactivation. J Exp Med 2007;204:595–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Zhu J, Peng T, Johnston C, Phasouk K, Kask AS, Klock A, et al. Immune surveillance by CD8αα+ skin-resident T cells in human herpes virus infection [published correction appears in Nature 2013;500:242] Nature 2013;497:494–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Zucman D, Truchis Pd, Majerholc C, Stegman S, Caillat-Zucman S. Prospective screening for human leukocyte antigen-B*5701 avoids abacavir hypersensitivity reaction in the ethnically mixed French HIV population. J Acquir Immune Defic Syndr 2007;45:1–3. [DOI] [PubMed] [Google Scholar]

RESOURCES