Next Article in Journal
Angiogenesis–Browning Interplay Mediated by Asprosin-Knockout Contributes to Weight Loss in Mice with Obesity
Next Article in Special Issue
Bacterial Motility and Its Role in Skin and Wound Infections
Previous Article in Journal
Allosteric Inhibition of c-Abl to Induce Unfolded Protein Response and Cell Death in Multiple Myeloma
Previous Article in Special Issue
The Microbiome in Systemic Sclerosis: Pathophysiology and Therapeutic Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Zinc and Zinc Transporters in Dermatology

1
Department of Applied Biotechnology, Ajou University, Suwon 16499, Republic of Korea
2
Department of Natural Sciences, Ajou University, Suwon 16499, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(24), 16165; https://doi.org/10.3390/ijms232416165
Submission received: 8 November 2022 / Revised: 9 December 2022 / Accepted: 13 December 2022 / Published: 18 December 2022
(This article belongs to the Special Issue Dermal Research: From Molecular Mechanisms to Pathology)

Abstract

:
Zinc is an important trace mineral in the human body and a daily intake of zinc is required to maintain a healthy status. Over the past decades, zinc has been used in formulating topical and systemic therapies for various skin disorders owing to its wound healing and antimicrobial properties. Zinc transporters play a major role in maintaining the integrity of the integumentary system by controlling zinc homeostasis within dermal layers. Mutations and abnormal function of zinc-transporting proteins can lead to disease development, such as spondylocheirodysplastic Ehlers–Danlos syndrome (SCD-EDS) and acrodermatitis enteropathica (AE) which can be fatal if left untreated. This review discusses the layers of the skin, the importance of zinc and zinc transporters in each layer, and the various skin disorders caused by zinc deficiency, in addition to zinc-containing compounds used for treating different skin disorders and skin protection.

1. Zinc Properties

Zinc is the second most abundant trace mineral in the human body [1]. It is a major constituent of every cell and is involved in cellular metabolic activities. The human body cannot make zinc on its own nor store it, therefore, a daily intake of zinc is required to maintain a steady status [2]. Recommended daily intake of zinc for an adult range from 8 to 11mg per day [3]. Zinc plays a major role in DNA synthesis. Zinc-deficient individuals are exposed to DNA damage; leading to impaired growth, delayed sexual maturation, hypogeusia, and hypogonadism [4]. Zinc supports immunity through maintaining Metallothionein (MT) homeostasis of the inflammatory response during aging [5], and insufficient expression of zinc-finger transcription factors in mRNA coding of growth factors leads to impaired wound healing [6]. Zinc also aids in the function of more than 200 enzymes by activating protein metabolism, in addition to protein synthesis [7,8]. As zinc enhances re-epithelialization, it reduces inflammation and bacterial growth [5]. Moreover, zinc supports normal fetal growth and development during pregnancy, childhood, and adolescence [9,10,11].

2. The Skin’s Anatomy and Physiology

The skin is the body’s largest organ [12], and it is made of fats, protein, water, and minerals. This integumentary system consists of three major layers: the outermost layer or the epidermis, the middle layer or the dermis, and the lowermost layer or the hypodermis. With each layer serving a different purpose, it all pours into protecting the body from microbes and other elements, providing a protective barrier against mechanical, thermal, and physical injury and hazardous substances, preventing loss of moisture, lowering the effects of ultraviolet (UV) rays, in addition to producing vitamin D [13,14] (Figure 1).

2.1. The Epidermis

The Epidermis is an avascular layer made of keratinized stratified squamous epithelium comprising four major layers, the stratum corneum, which is the most superficial layer of the epidermis, is a thick layer made up of dead keratinocytes known as corneocytes [15]. Corneocytes protect the skin from injuries, UV light, and microbes [16]. This layer also protects the skin from losing its water content. Deep to the stratum corneum is the stratum lucidum [17], which is a thin layer made of flattened corneocytes that is present on palms and soles [18]. Beneath the stratum lucidum, is the stratum granulosum, which consists of keratinocytes preparing to become flattened corneocytes [19]. Stratum spinosum lays down to the stratum granulosum and contains mature keratinocytes with visible granules that adhere to each other by desmosomes and produce keratin, a protein that helps form hair and nails [20,21]. The deepest layer of the epidermis is the stratum basale or stratum germinativum. This layer contains new keratinocytes in their developing stage, Merkel cells, and stem cells [22]. This layer contains melanocytes that produce melanin responsible for pigmentation.

2.2. The Dermis

The dermis is the middle layer of the skin and it is composed of fibrous, filamentous, and amorphous connective tissue [23]. This layer allows stimuli inductions to control entry by the vascular and neural networks, epidermal appendages, fibroblasts, mast cells, macrophages, and other blood cells and it also accommodates sweat glands, sebaceous glands, and hair follicles in addition to allowing substance exchange to both the dermis and the epidermis through the epidermal–dermal junction [24]. The components of the dermal layer, such as collagen, can also vary depending on external stimuli [25]. Collagen is a structural protein that is considered a principal component of the dermal layer with at least 28 genetically different types [26], making up 70% to 80% of the skin’s dry weight [27]. This stress-resistant material is degenerated by spare collagenases [28]. The dermis can be divided into two main regions, the upper papillary layer, and the lower reticular layer [29]. In the papillary and adventitial dermis, Collagen type I fibers are loosely positioned, whereas in the reticular dermis, are present in bulky bundles [29,30]. In the basement membrane, collagen type IV is present, while keratinocytes mainly produce collagen type VII [31].
The dermal vasculature is made of two interrelated plexuses: the superficial subpapillary plexus branches into capillaries that extend to papillae supplying the epidermis, and the deep vascular plexus that is supplied by larger blood vessels extending from the hypodermis [32]. In addition to vascular networks surrounding sweat glands and hair follicles in the dermis, the papillary layer embodies muscle fibers of the arrectores pili that are attached to the hair follicle causing it to be pulled vertically upon contraction resulting in a “goose-bumps” skin condition [33]. The sensation of pain, temperature, and itchiness is attributed to the presence of unmyelinated nerve fibers ending around hair follicles and the papillary layer while mediating touch is regulated by Meissner corpuscles that are mostly present on the palms and soles with more concentration in the fingertips [34,35]. The sensation of pressure is attributed to Pacini corpuscles that are present on the weight-bearing surfaces, genital, nipple, and anogenital reticular dermis [36]. Post-ganglionic adrenergic fibers regulate vasoconstriction through the secretions of apocrine gland secretions and the arrectores pili muscle contractions while the cholinergic fibers regulate eccrine sweat gland secretions [37]. Mast cells are linked to allergic reactions, and they accumulate in large numbers in the papillary dermis and are also found in the subcutaneous fat [38]. Allergens activate mast cells leading to the release of cytokines and chemokines that are responsible for initiating an inflammatory reaction [39].

2.3. The Hypodermis

The lowermost layer of the integumentary system, also known as the subcutaneous tissue, is composed of mainly connective tissue and adipose tissue, and it lies between the dermis and the skeletal muscle [40]. The hypodermis contains adipocytes, large blood vessels, nerves, and adipose stromal cells and it is important for thermoregulation, and protection of the underlying structures from traumas, energy reservation, and hormone production (leptin) in addition to supporting keratinocyte and fibroblast proliferation [41]. The deposition of adipocytes in this layer depends on age, hormones, genetic factors, and body region [42,43,44], while the size of adipocytes depend on nutrition [45]. Other components of the hypodermis are macrophages, fibrous bands, collagen, and elastin that connect the subcutaneous layer to the dermis, whereas hair follicle roots can also be found embedded in the hypodermis [46].

3. Zinc Distribution in the Human Skin

Zinc is absorbed in the small intestines through a carrier-mediated mechanism [47], and it is distributed to the rest of the body in different amounts. The vast majority goes to the skeletal muscle (60%), followed by the bone (30%) and the skin (5%), the liver (5%), and other organs (2–3%) [48,49,50]. Different illnesses and high phytate-containing foods can inhibit zinc uptake thereby hindering its bioavailability in the body [3]. The skin contains approximately 60 μg/g of zinc in the epidermis and 40 μg/g in the upper dermis [51]. Zinc deficiency can be first exhibited on the skin causing skin diseases such as acrodermatitis enteropathica [3,52].

4. Role of Zinc in the Skin

Zinc can bind ~10% of the total proteins identified in the human body, according to bioinformatics research on the human genome. Zinc concentration was found higher in the stratum spinosum than in other epidermal layers [23]. While in the dermis, zinc can be found in higher concentrations in the upper dermis than the lower dermis due to higher mast cell accumulation that is rich in zinc content necessary for cytokine production and FcεRI-dependent degranulation. It was observed that the maintenance of adequate zinc levels within cultured HaCat keratinocytes promotes the survival and proliferation of these cells while zinc deficiency causes activation of a DNA fragment and caspase-3 inducing apoptosis [53]. Extracellular Zn2+ is released following skin injury causing activation of the G-protein coupled receptor (GPR39) and zinc-sensing receptor (ZnR) pathway that is expressed in the keratinocytes and other epithelial cells, leading to the repair of the epithelium. Zinc possesses anti-inflammatory properties in suppressing the generation of inflammatory cytokines [54], and the liberation of zinc ions from zinc oxide (ZnO) nanomaterials aids in wound healing [55], therefore, it has been widely utilized in formulating cosmetics and ointments [56,57]. ZnO nanoparticles and Zn2+ ions exhibit different spectrums of antimicrobial activities [58,59,60,61,62], and it shows more potent antibacterial properties when combined with chitosan hydrogel, making it a great component for wound dressings [63]. Zinc deficiency impairs the proliferation, differentiation, survival of keratinocytes, and wound healing, in addition to increasing the production of ATP, inflammatory cytokines, and iNOS/NO by keratinocytes in addition to causing a telogen effluvium and an abnormal hair keratinization [49,64]. A less toxic zinc-based metal–organic framework (Zn-BTC), with the ability to slowly release Zn2+, was found to exhibit biocompatibility, antibacterial and anti-inflammatory properties. It aids the skin’s wound healing process as Zn-BTC lowers the expression of certain antioxidant genes and enhances the expression of wound healing genes, in addition to, exhibiting better bactericidal effect on different drug-resistant bacteria through reducing 41.4% MRSA and 47.2% Escherichia coli in rats [62]. It was revealed that combining polyethylene glycol (PEG) and ZnO nanoparticles into an FDA-approved bioabsorbable polyester, Poly 4-hydroxybutyrate (P4HB)’s matrix resulted in Zn2+ ion release, which allowed for better blood clotting, better bacterial elimination, prevention of bacterial adhesion, and had as well demonstrated excellent hemostatic performance [65]. With the ongoing advancement of nanotechnology in drug delivery, a recent study has confirmed the potential of ZnO nanoparticles in wound healing through the synthesis of LED illuminated curcumin loaded Zinc Oxide (Cu + ZnO NP) nanoparticles that demonstrated an enhanced wound contraction, collagen deposition, angiogenesis, and re-epithelialization, by which in turn, accelerated the overall wound healing process [66]. For skin burns, zinc silicate nanoparticles-based scaffolds were found to enhance Human Umbilical Vein Endothelial Cells (HUVECs) angiogenic activity and Schwann cells’ neurogenic activity, as well as displaying remarkable blood vessel and nerve fiber regeneration abilities, both of which are required for effective skin tissue regeneration, thereafter, it enables a better the healing of innervated and vascularized skin burn wounds [67]. As zinc is required for the integrity of the integumentary tissue, degradation of zinc levels in the skin can lead to a variety of diseases, some are inherited while others are acquired through low dietary intake while other zinc deficiency conditions are linked to intestinal malabsorption [4,68].

5. Role of Zinc Transporters in the Skin

Zinc transporters are membrane proteins that control the transportation and concentration of zinc both inside the cellular compartments and outside the cells by regulating the influx and efflux of zinc, which is crucial in maintaining homeostasis within the tissues [48]. Primary zinc transporters include the zinc transporter ZnT (SLC30) family and the Zrt/Irt-like protein/solute carrier family ZIP (SLC39) [69,70]. These transporters directly influence zinc availability and pathogenesis. There are ten members of the zinc efflux transporter family (ZnT) and fourteen members of the zinc inflow transporter family (ZIP) that have been identified in mammals [70,71]. According to research, ZnT transporters and their homologs function as Zn2+/H+ antiporters [72], while ZIP transporters act as a symporter for zinc and other metals/bicarbonates [73,74]. A study suggested that ZIP transporters act as a zinc-selective channel that transports zinc ions into the cytoplasm based on zinc concentration gradients [75]. ZIP family was found to be directly involved in maintaining skin homeostasis [76]. In the epidermis, ZIP1, ZIP2, ZIP4, and ZIP10 are linked to epidermal morphogenesis and abnormalities [23,77,78,79], whereas ZIP7 and ZIP13 are required for normal dermal development and collagen metabolism [80]. Although zinc transporters play a major role in maintaining cellular homeostasis, metallothioneins (MT) accumulating in the epidermal layer, binding heavy metal ions such as zinc are found to be associated with increased zinc concentrations in the tissues which implies the significance of not only ZIP but also MTs in maintaining high concentrations of zinc required for normal proliferation and differentiation of the epidermis [76,81]. Acrodermatitis enteropathica (AE) is a zinc metabolism disorder that can be inherited or acquired, and it is caused by dysfunction of ZIP4 protein leading to impaired zinc absorption [82]. Spondylocheiro dysplastic form of Ehlers–Danlos syndrome (SCD-EDS) is an autosomal-recessive entity that is initiated by mutation of the zinc transporter ZIP13 causing hyperplasia and weakening of the skin and hypermobility in the small joints [83]. Epidermodysplasia verruciformis (EV) is an autosomal-recessive skin disease associated with a high risk of skin cancer [84,85] developing in individuals vulnerable to specific genotypes of human papillomavirus, such as the oncogenic HPV-5. EV patients show mutations in EVER1 or EVER2 genes that form a complex with ZnT1 majorly in the endoplasmic reticulum of keratinocytes leading to an increase in free zinc transportation into the nucleus, and thus, promoting AP-1 activity that causes abnormal replication of EV-related oncogenic HPVs responsible for skin cancer development [49,86]. Poor secretion of zinc into breast milk causes a condition called Transient Neonatal Zinc Deficiency resulting from ZnT2 mutation as ZnT2 transports zinc from the cytoplasm to cytoplasmic secretory vesicles [87,88]. ZIP2 aids in the differentiation of keratinocytes in the epidermis and the knock-down of ZIP2 leads to immortalizing human keratinocytes and inhibiting their differentiation [23] (Figure 2).

6. Function of Zinc, and Zinc Transporters in Dermal Skin Cells

6.1. Function of Zinc in the Dermal Layer

As the dermal layer consists of connective tissue containing nerve endings, sweat glands, oil glands, and hair follicles, zinc promotes dermal homeostasis through zinc transporters and MT proteins to regulate the levels of zinc across body cells’ phospholipid bilayers [8,76]. Zinc has anti-inflammatory properties and influences main pro-inflammatory signaling pathways as in down-regulating inflammatory cytokines release [89].

6.2. Function of Zinc Transporters in the Dermal Layer

There are 14 identified zinc transporters within the ZIP family [70]. Among them, ZIP7 (SLC39A7) and ZIP13 (SLC39A13) zinc-regulating transporters are required for optimal development of the dermal layer [80]. These transporters regulate cytosolic zinc levels by delivering the required zinc quantities for optimal function [90]. ZIP7 transports zinc from the endoplasmic reticulum (ER) stores to the cytoplasm and regulates zinc homeostasis in the Golgi apparatus [91,92]. Whereas ZIP13, a transporter protein required for connective tissue development [93], functions in transporting zinc from the vesicular stores to the ER and other compartments [94]. ZIP13 dysfunction leads to a decrease in zinc levels in the cytoplasm and ER, causing ER dysfunction and stress dysfunction. Thereafter, any abnormalities within these transporters can lead to improper dermal formation, thus causing dermal impairments such as dermal dysplasia [92,95]. Transforming growth factor beta (TGF-β)–SMAD–ZIP13 axis was also found to be necessary for dermal formation [92].

6.3. ZIP7 Transporter

ZIP7 is found in the ER promoting zinc homeostasis [80]. However, recent studies suggest that ZIP7 can also be present in the Golgi apparatus [91]. ZIP7 is involved in the formation of the skin’s connective tissue. Unlike other members of the SLC39A family, ZIP7 has a histidine binding region known as N-termini that acts as a zinc ligand maintaining homeostasis within the cell’s ER lumen where ZIP7 resides along with labile zinc [96]. ZIP7-knockout mice showed reduced dermal and hypodermal thickness, indicating ZIP7 is required for connective tissue development [92]. ZIP7-knockout leads to decreased cell density, thereby, thinner connective tissue indicating that ZIP7 is crucial for human mesenchymal stem cell (hMSC) proliferation [92]. ZIP7 is more predominant in the hMSCs than ZIP13 and is found to be essential in fibrogenic and osteogenic development suggesting ZIP7 is required for preventing ER stress and thus, preservation of hMSCs [80].
Contrary to ZIP13, the knockdown of ZIP7 in cells exhibits enhanced ER stress, accompanied by higher zinc concentrations and aggregation of protein disulfide isomerase (PDI) leading to unfolded protein response, suppressing cell growth and eventually, apoptosis [76].
In pre-B and immature B cells, ZIP7 is necessary for promoting gene transcription affecting the normal transition from late pre-B to immature B cells. In activated human primary B cells, about 50% ZIP7-containing compartments were found within 1 µm of the plasma membrane while more widespread distribution was observed in the cytoplasm of considerably bigger HEK293T cells. This finding indicates that alterations in local or dynamic ZIP7 dissemination of Zn2+ can vary by cell type [97]. ZIP7 deficiency stimulates the death of normally developing B cells, disrupting its differentiation process as it fundamentally modifies the gene expression program according to RNA sequencing. The decreased cytoplasmic Zn2+ associated with ZIP7 deficiency is expected to result in pathologically elevated phosphatase activity and, as a result, contribute to impaired pre-BCR- and BCR-dependent signaling at the positive selection checkpoints. By employing flow cytometry and a cell-permeable alkaline phosphatase substrate, it was discovered that ZIP7 had greater constitutive phosphatase activity than WT pre-B and immature B cells.

6.4. ZIP13 Transporter

ZIP13 regulates zinc homeostasis in the Golgi apparatus [80]. ZIP13 is a homo-dimerized zinc transporter containing eight transmembrane domains with N and C termini in the hydrophilic region [95]. The expression of Drosophila ZIP13 (dZIP13) in Saccharomyces cerevisiae revealed that it is mainly regulated by iron abundance in protein level with a slight iron response within mRNA as the presence of iron in excess amounts activates iron binding affinity boosting its efflux and preventing dZIP13 from deterioration [98].
It was found that the transforming growth factor beta (TGF-β) has a positive correlation with the mRNA expression of ZIP13 and contributes to the differentiation of beige adipocytes by regulating C/EBP-β protein levels [80,99]. ZIP13 deficiency accelerates adipocyte browning of the adipose tissue as C/EBP-β leads to adipogenesis and brown adipocyte differentiation [99]. As ZIP13 histamine residues are required for ZIP13-mediated zinc transport to suppress adipocyte browning, the inadequacy of this transporter causes an increase in C3H10T1/2 cells that can differentiate into beige or brown adipocytes when a brown adipogenic cocktail is administered [93,100]. It is also suggested that the interrupted zinc transportation from zinc stores [94], to the cytosol causes ER stress, leading to the development of Ehlers–Danlos syndrome, spondylodysplastic type 3 (EDSSPD3) [76,94]. The nuclear translocation of SMAD transcription factors in the BMP/TGF signaling pathway was reduced in ZIP13-knockout mice with phosphorylation status remaining unchanged, indicating that ZIP13 deficiency causes an impairment in BMP/TGF-β signaling pathway leading to abnormally shaped collagen-producing cell, shrinking cartilage, and poor chondrocyte differentiation, resulting in decreased collagen synthesis [76,93].
ZIP13 is also distributed in the intracellular vesicles [94]. Many enzymes requiring zinc are found in the secretory pathway, including Calnexin (Cnx) and calreticulin (Crt) in the ER, and glycosylphosphatidylinositol (GPI) phosphoethanolamine transferase and other zinc-secreted enzymes including matrix metalloproteases, alkaline phosphatases (ALP), and angiotensin-converting enzymes. ALPs can be an indicator of zinc deficiency in the ER and Golgi as they require zinc for their activity [94].

6.5. Analogies of ZIP7 and ZIP13

There are two sets of differentially expressed genes (DEGs) of both ZIP7 knockdown (KD) and ZIP13-KD in hMCS that overlap, indicating ZIP7 and ZIP13 share functional characteristics through the overexpression of shared genes [80].
ZIP7 and ZIP13 were found to have functional distinctiveness and similarities. ZIP7-KD and ZIP13-KD both upregulated ER stress-related genes, genes enriched in cysteine-type endopeptidase, wnt signaling pathway mechanisms, and blood coagulation. In addition, downregulated proliferation-related genes and genes enriched in glucocorticoid and hyp zip7 and zip13oxia responses as well as mRNA splicing processes. The downregulated genes were implicated in inflammatory and immunological responses indicating that both ZIP7-KD and ZIP13-KD are crucial for the immune response of hMSCs in either zinc-dependent and/or zinc-independent way.
Both ZIP7 and ZIP13 are equally necessary for collagen synthesis, as it was found that the knockout of ZIP7, with collagen promoter in control of Cre-Lox recombination, causes connective tissue disorders, corresponding to that of Zip13-knockout mice [76].

7. Zinc Deficiency-Related Skin Disorders

Approximately one-third of the world’s population suffers from zinc deficiency following a consequence of low zinc consumption, malabsorption, or increased loss. Low consumption of zinc is endemic to Southeast Asia, sub-Saharan Africa, rural Iran, Turkey, and Egypt [101,102]. Other zinc deficiency predisposing factors include a low-protein diet, a vegetarian diet, eating disorders such as anorexia nervosa or bulimia nervosa, parenteral nutrition, hookworm infection, AE, formula milk low in zinc, and other gastrointestinal and renal dysfunctions. Other zinc deficiency-related skin disorders include necrolytic migratory erythema [103,104], pellagra [105], and pressure ulcers [106].
In infants, zinc deficiency is attributed to either classic acrodermatitis enteropathica, maternal milk low on zinc, or premature infants with prolonged parenteral nutrition, all of which can be reversed upon zinc supplementation [107,108].

7.1. Acrodermatitis Enteropathica (AE)

Acrodermatitis is a zinc deficiency skin disease that is either hereditary or acquired [109]. It is attributed to zinc malabsorption in the duodenum or maldistribution of zinc to bodily tissues [110,111]. The latter can be diagnosed by identifying lower-than-normal ALP levels in blood serum [82]. Some studies suggested that the malabsorption of zinc is caused by a defect in the pancreatic zinc-binding ligands that transport zinc [112,113].
AE is characterized by the presence of cutaneous lesions that vary from mild to severe and are located around body openings and nails, with mild or severe diarrhea and alopecia [110,114]. Secondary infections due to immunosuppression are common and if AE is left untreated it can lead to death. Another form of AE showing zinc deficiency symptoms is observed in infants before weaning occurring as a result of reduced zinc secretion into the mother’s milk [115,116]. The identification of AE is performed by detecting low plasma zinc levels and it can be reversed with proper zinc supplementation [109].

7.2. Pathogenesis of AE

Zinc deficiency leads to elevating ATP and ADP levels and reducing adenosine levels in all cells due to extracellular adenine-nucleotide hydrolysis suppression in addition to causing a decrease in the activity of four major ectoenzymes (ENPP1, ENPP3, NT5E/CD73, and TNAP involved in the hydrolysis of extracellular ATP to adenosine through ADP and AMP). As a result, zinc affects extracellular adenine-nucleotide metabolism, and its deficiency slows both extracellular ATP clearance and adenosine production [117,118]. ZnT zinc transporters ZnT5 and ZnT6 heterodimers and ZnT7 homodimers may also increase vulnerability to zinc deficiencies as they are both required for TNAP and NT5E/CD73 activity [117,119]. It is also suggested that zinc deficiency causes a decrease in Langerhans cells that express ENTPD1/CD39 leading to severe acrodermatitis [120,121,122,123].

7.3. Spondylocheirodysplastic Ehlers–Danlos Syndrome (SCD-EDS)

EDS is an inherited disorder of connective tissue [124]. A new type of EDS was reported with slightly different features from that of the typical EDS [83]. The connective tissue disorders include hyperplastic skin, and articular hypermobility, while physical signs include wrinkled palms, short stature, tapered fingers, absence of periorbital tissue, and antimongoloid slant [125]. Similar clinical signs were observed in Zip13-KO mice, including impaired cartilage development, growth retardation, kyphosis, osteopenia, and craniofacial abnormalities with a reduction in corneal and dermal stromal collagen levels. SCD-EDS is attributed to a mutation in the ZIP13 protein of the LIV-1 subfamily [83].

7.4. Pathogenesis of SCD-EDS

Knocking out ZIP13 in mice revealed maturation defects in cells originating in the mesenchyme which led to retardation in connective tissue development [93]. Analysis revealed that ZIP13-KO cells demonstrate an impaired nuclear translocation of the transcription factor SMADs, responding to BMP and TGF-β necessary for connective tissue development [126,127]. An increase in zinc levels in the Golgi apparatus with a decrease in the nucleus was detected in Zip13-KO cells indicating a disruption in intracellular zinc homeostasis [83].
Therapeutic strategy for SCD-EDS can be achieved through restoring intracellular zinc homeostasis [94], and the removal of mutant ZIP13 protein via the ubiquitin-proteasome pathway [83]. VCP and HSP90 molecules are involved in the unfolding and transport of mutant ZIP13 to the proteasome. Inhibiting these molecules results in mutant ZIP13 protein accumulation within cells. The pathogenic ZIP13 mutants’ characteristics could be easily reversed by the proteasome inhibitors MG-132 and lactacystin [128]. These inhibitors are toxic, activating certain signaling pathways that can lead to cell death. Proteasomes take part in a variety of biological activities, such as cell proliferation, gene regulation, stress response, and apoptosis [129,130]. Therefore, proteasome-dependent degradation causes high cell toxicity [131,132,133]. For that, finding treatment remains challenging. Bortezomib is an example of a drug that was approved for human use but was later found to be causing cytotoxicity over the long term [134,135]. Proteasomes require protein-degradation folding molecules known as chaperones for the degradation of the cell membrane [136,137]. These chaperones can be targeted instead of proteasomes for eliminating pathogenic ZIP13 mutant protein in the treatment of SCD-EDS as they are not usually necessary for cell survival. Chaperone inhibitors, DBeQ and 17AAG were found to trigger the buildup of pathogenic ZIP13 mutant protein and restore zinc homeostasis within the cell [138]. Modification of chaperone inhibitors such as DBeQ and 17AAG to minimize cell toxicity can be an effective therapeutic strategy in the treatment of SCD-EDS [83].

8. Zinc in the Therapy of Skin Disorders

In dermatology, the topical application of zinc has been utilized widely in the treatment of various skin diseases of different etiologies as zinc possesses anti-bacterial and anti-inflammatory properties as well as offering photoprotection without causing adverse effects [5,6,139].

8.1. Acne Vulgaris

There are few theories that explain the pathogenesis of acne vulgaris [140,141,142,143]. In general, androgens and hyperkeratinization of the skin cause obstruction of sebaceous glands, thus, allowing the proliferation of Propionibacterium acnes bacteria. The latter causes inflammatory cells to gather at the site leading to metabolization of sebum’s triglyceride that forms free fatty acids and inflammatory mediators complex resulting in irritation.
Treatment protocols have been designed to tackle different stages in the pathogenesis of acne. These treatment methods can either be topical or systemic. Topical treatments include using wash gels and lotions or antibiotics, while systemic treatment includes the use of retinoids, hormonal mediators, or antibiotics. Both treatment methods have versatile side effects on the outer skin and internal organs, such as erythema, dryness, peeling, teratogenicity, high risks of embolism, and the development of antimicrobial resistance [144]. Through several clinical trials, it has been proved that zinc has the potential to reduce acne by inhibiting P. acnes proliferation, suppressing sebaceous gland activity, regulating DNA and RNA polymerases, and gene transcription [145,146,147]. It was found that combining topical zinc with erythromycin is superior to topical clindamycin or erythromycin alone in treating acne and is of equal effect with tetracycline [148]. It also showed an earlier onset of action compared to conventional treatment methods. Nels®, a zinc oxide-containing cream, was found to improve acne equally to benzoyl peroxide with fewer side effects [149]. Oral zinc or zinc plus oral vitamin A was found to outdo vitamin A alone [150,151].

8.2. Zinc in the Treatment of Other Skin Disorders

Various skin disorders such as psoriasis and eczema can be successfully managed with zinc-containing compounds as shown in (Table 1).

9. Cosmeceutical Application of Zinc

Exposure to UV harmful sunrays of 320–400 nm (UVA) and 290–320 nm (UVB) can pose potential threats to the health and integrity of the skin’s connective tissue through initiating abnormalities such as melasma, variety of skin cancers, and photoaging. UVA was found to generate free radicals causing photoaging in human skin as well as promoting carcinogenesis and immunosuppression while UVB was found to be responsible for causing squamous cell carcinoma in animals [227,228,229,230].
As a UV blocking agent, FDA-approved ZnO has been widely employed in the formulation of inorganic (physical) sunscreens as a nonirritating, insoluble agent that can absorb, scatter, and reflect UV radiation of 290–380 nm responsible for triggering severe oxidative stress leading to DNA damage and apoptosis [231,232,233]. As a sunscreen component, microfine ZnO was found to outperform its microfine titanium dioxide alternative in providing better protection against long-wave UVA and in looking less white on the skin [234].
Initial sunscreen preparations lacked aesthetic appeal due to ZnO’s chalky white textural properties [235,236]. As the evolution in nanotechnology has emerged, ZnO particles were reduced to nanoparticles of less than 100 nm, which helped lessen the white chalky appearance. Generally, reduction in particle size has given rise to toxicity and oxidative stress concerns as well [237]. In the case of coated and noncoated ZnO-NP, it did not permeate the stratum corneum or cause local skin toxicity after an in vivo 5-day trial [238]. A large body of evidence supports the safe use of ZnO in sunscreen preparations and non-sunscreen preparations for the treatment of various skin disorders [6,139,238,239,240].
Since the human body has high levels of endogenous zinc, minimal transdermal absorption may not pose any adverse health effects as ZnO particles would dissociate into zinc and oxygen ions, both of which exist naturally in the human body [241]. The body’s homeostatic system controls the uptake, distribution, and excretion of zinc [242]. Respectively, oxygen absorption through the skin is safe and essential for life. The recommended ZnO concentration in sunscreen preparations should be no more than 25% according to the European Commission’s Scientific Committee on Consumer Safety [243].
Active ingredients reaching the viable layers of the skin used in combination with ZnO or Titanium Oxide in sunscreen preparations should be further investigated for their safe use as they can impact reproduction, development, or carcinogenesis [244].
As a waste material, a study evaluating the safety of anthropogenic inorganic sunscreen filters in seawater waste on coral reefs revealed that uncoated ZnO nanoparticles can cause severe, rapid bleaching of Acropora spp. [245].

10. Conclusions

Zinc is a trace element that plays a major role in maintaining healthy status since early infancy until elderliness. Subsequently, changes in required zinc levels can result in different dermatological disorders that can mostly be reversed with adequate zinc supplementation, while the involvement of abnormally functioning zinc transporters can limit the efficacy of zinc supplementation. Zinc-containing compounds remain a favorable therapeutic option for various dermatological disorders due to its lack of serious side effects upon topical use. Zinc and zinc transporters have been extensively investigated in the past few decades, but a large body of evidence is still missing such as a clear understanding of precise subcellular zinc transportation following cellular stimuli, each zinc transporter structure and their efflux and influx mechanisms, or the identification of the specific components mediated by zinc and their functions in protein networking. In the formulation of sunscreens, ZnO’s UV rays scattering and reflecting properties allowed for its extensive and safe use on daily basis, but the safety of chemical UV-filters used in combination with ZnO require further investigations.

Author Contributions

Conceptualization, Z.A.-K., S.B., and B.-H.B.; Writing—Original Draft, Z.A.-K.; Writing—Review and Editing, S.B.; Supervision, B.-H.B.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the National Research Foundation of Korea (NRF) funded by the Korean government (MSIT) (No. 2019005607 to B.-H.B.), the Ajou University Research Fund (to B.-H.B.), a grant provided by the Korea Initiative for fostering University of Research and Innovation Program of the National Research Foundation (NRF) funded by the Korean government (MSIT) (No. NRF2021M3H1A104892211; to B.-H.B).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Silva, C.S.; Moutinho, C.; Ferreira da Vinha, A.; Matos, C. Trace minerals in human health: Iron, zinc, copper, manganese and fluorine. Int. J. Sci. Res. Methodol. 2019, 13, 57–80. [Google Scholar]
  2. Vollmer, D.L.; West, V.A.; Lephart, E.D. Enhancing skin health: By oral administration of natural compounds and minerals with implications to the dermal microbiome. Int. J. Mol. Sci. 2018, 19, 3059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Roohani, N.; Hurrell, R.; Kelishadi, R.; Schulin, R. Zinc and its importance for human health: An integrative review. J. Res. Med. Sci. 2013, 18, 144. [Google Scholar] [PubMed]
  4. Prasad, A.S. Discovery of human zinc deficiency: Its impact on human health and disease. Adv. Nutr. 2013, 4, 176–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lin, P.-H.; Sermersheim, M.; Li, H.; Lee, P.H.U.; Steinberg, S.M.; Ma, J. Zinc in Wound Healing Modulation. Nutrients 2018, 10, 16. [Google Scholar] [CrossRef] [Green Version]
  6. Lansdown, A.B.G.; Mirastschijski, U.; Stubbs, N.; Scanlon, E.; Ågren, M.S. Zinc in wound healing: Theoretical, experimental, and clinical aspects. Wound Repair Regen. 2007, 15, 2–16. [Google Scholar] [CrossRef]
  7. MacDonald, R.S. The role of zinc in growth and cell proliferation. J. Nutr. 2000, 130, 1500S–1508S. [Google Scholar] [CrossRef] [Green Version]
  8. Jarosz, M.; Olbert, M.; Wyszogrodzka, G.; Młyniec, K.; Librowski, T. Antioxidant and anti-inflammatory effects of zinc. Zinc-dependent NF-κB signaling. Inflammopharmacology 2017, 25, 11–24. [Google Scholar] [CrossRef] [Green Version]
  9. Gluckman, S.P.; Hanson, M.; Seng, C.Y.; Bardsley, A.; Gluckman, P.; Hanson, M.; Seng, C.Y.; Bardsley, A. 196Zinc in pregnancy and breastfeeding. In Nutrition and Lifestyle for Pregnancy and Breastfeeding; Oxford University Press: Oxford, UK, 2014; p. 196. [Google Scholar]
  10. Ota, E.; Mori, R.; Middleton, P.; Tobe-Gai, R.; Mahomed, K.; Miyazaki, C.; Bhutta, Z.A. Zinc supplementation for improving pregnancy and infant outcome. Cochrane Database Syst. Rev. 2015, 2015, Cd000230. [Google Scholar] [CrossRef]
  11. Glutsch, V.; Hamm, H.; Goebeler, M. Zinc and skin: An update. JDDG J. Dtsch. Dermatol. Ges. 2019, 17, 589–596. [Google Scholar] [CrossRef] [Green Version]
  12. Liu, Y.; Pharr, M.; Salvatore, G.A. Lab-on-skin: A review of flexible and stretchable electronics for wearable health monitoring. ACS Nano 2017, 11, 9614–9635. [Google Scholar] [CrossRef] [PubMed]
  13. Jablonski, N.G. Skin: A Natural History; University of California Press: Berkeley, CA, USA, 2008. [Google Scholar]
  14. Holick, M.F. Sunlight, UV-radiation, vitamin D and skin cancer: How much sunlight do we need? In Sunlight, Vitamin D and Skin Cancer; Springer: New York, NY, USA, 2008; pp. 1–15. [Google Scholar]
  15. Gilaberte, Y.; Prieto-Torres, L.; Pastushenko, I.; Juarranz, Á. Chapter 1—Anatomy and Function of the Skin. In Nanoscience in Dermatology; Hamblin, M.R., Avci, P., Prow, T.W., Eds.; Academic Press: Boston, MA, USA, 2016; pp. 1–14. [Google Scholar]
  16. Biniek, K.; Levi, K.; Dauskardt, R.H. Solar UV radiation reduces the barrier function of human skin. Proc. Natl. Acad. Sci. USA 2012, 109, 17111–17116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Yousef, H.; Alhajj, M.; Sharma, S. Anatomy, Skin (Integument), Epidermis; StatPearls Publishing: Treasure Island, FL, USA, 2017. [Google Scholar]
  18. Murphrey, M.B.; Miao, J.H.; Zito, P.M. Histology, Stratum Corneum; StatPearls Publishing: Treasure Island, FL, USA, 2018. [Google Scholar]
  19. Del Rosso, J.Q.; Levin, J. The clinical relevance of maintaining the functional integrity of the stratum corneum in both healthy and disease-affected skin. J. Clin. Aesthetic Dermatol. 2011, 4, 22–42. [Google Scholar]
  20. Agarwal, S.; Krishnamurthy, K. Histology, Skin; StatPearls Publishing: Treasure Island, FL, USA, 2019. [Google Scholar]
  21. McKittrick, J.; Chen, P.-Y.; Bodde, S.; Yang, W.; Novitskaya, E.; Meyers, M. The structure, functions, and mechanical properties of keratin. JOM 2012, 64, 449–468. [Google Scholar] [CrossRef]
  22. Hou, J. Chapter 7—Paracellular Channel in Organ System. In The Paracellular Channel; Hou, J., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 93–141. [Google Scholar]
  23. Inoue, Y.; Hasegawa, S.; Ban, S.; Yamada, T.; Date, Y.; Mizutani, H.; Nakata, S.; Tanaka, M.; Hirashima, N. ZIP2 protein, a zinc transporter, is associated with keratinocyte differentiation. J. Biol. Chem. 2014, 289, 21451–21462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Briggaman, R.A.; Wheeler, C.E. The Epidermal-Dermal Junction. J. Investig. Dermatol. 1975, 65, 71–84. [Google Scholar] [CrossRef] [Green Version]
  25. Venus, M.; Waterman, J.; McNab, I. Basic physiology of the skin. Surgery 2010, 28, 469–472. [Google Scholar]
  26. Ricard-Blum, S. The collagen family. Cold Spring Harb. Perspect. Biol. 2011, 3, a004978. [Google Scholar] [CrossRef] [Green Version]
  27. Oikarinen, A. Aging of the skin connective tissue: How to measure the biochemical and mechanical properties of aging dermis. Photodermatol. Photoimmunol. Photomed. 1994, 10, 47–52. [Google Scholar]
  28. Alipour, H.; Raz, A.; Zakeri, S.; Dinparast Djadid, N. Therapeutic applications of collagenase (metalloproteases): A review. Asian Pac. J. Trop. Biomed. 2016, 6, 975–981. [Google Scholar] [CrossRef] [Green Version]
  29. Brown, T.M.; Krishnamurthy, K. Histology, Dermis; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  30. Meigel, W.N.; Gay, S.; Weber, L. Dermal architecture and collagen type distribution. Arch. Dermatol. Res. 1977, 259, 1–10. [Google Scholar] [CrossRef] [PubMed]
  31. Chung, H.J.; Uitto, J. Type VII collagen: The anchoring fibril protein at fault in dystrophic epidermolysis bullosa. Dermatol. Clin. 2010, 28, 93–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Braverman, I.M. The Cutaneous Microcirculation. J. Investig. Dermatol. Symp. Proc. 2000, 5, 3–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Torkamani, N.; Rufaut, N.W.; Jones, L.; Sinclair, R.D. Beyond goosebumps: Does the arrector pili muscle have a role in hair loss? Int. J. Trichology 2014, 6, 88–94. [Google Scholar] [CrossRef]
  34. Ringkamp, M.; Schepers, R.J.; Shimada, S.G.; Johanek, L.M.; Hartke, T.V.; Borzan, J.; Shim, B.; LaMotte, R.H.; Meyer, R.A. A role for nociceptive, myelinated nerve fibers in itch sensation. J. Neurosci. 2011, 31, 14841–14849. [Google Scholar] [CrossRef] [Green Version]
  35. Piccinin, M.A.; Miao, J.H.; Schwartz, J. Histology, Meissner Corpuscle; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  36. Kolarsick, P.A.; Kolarsick, M.A.; Goodwin, C. Anatomy and physiology of the skin. J. Dermatol. Nurses’ Assoc. 2011, 3, 203–213. [Google Scholar] [CrossRef] [Green Version]
  37. James, W.D.; Elston, D.; Berger, T. Andrew’s Diseases of the Skin E-Book: Clinical Dermatology; Elsevier Health Sciences: Amsterdam, The Netherlands, 2011. [Google Scholar]
  38. Altintas, M.M.; Azad, A.; Nayer, B.; Contreras, G.; Zaias, J.; Faul, C.; Reiser, J.; Nayer, A. Mast cells, macrophages, and crown-like structures distinguish subcutaneous from visceral fat in mice. J. Lipid Res. 2011, 52, 480–488. [Google Scholar] [CrossRef] [Green Version]
  39. Galli, S.J.; Tsai, M.; Piliponsky, A.M. The development of allergic inflammation. Nature 2008, 454, 445–454. [Google Scholar] [CrossRef] [Green Version]
  40. Lopez-Ojeda, W.; Pandey, A.; Alhajj, M.; Oakley, A.M. Anatomy, Skin (Integument); StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  41. Wong, R.; Geyer, S.; Weninger, W.; Guimberteau, J.-C.; Wong, J.K. The dynamic anatomy and patterning of skin. Exp. Dermatol. 2016, 25, 92–98. [Google Scholar] [CrossRef] [Green Version]
  42. Rivera-Gonzalez, G.; Shook, B.; Horsley, V. Adipocytes in skin health and disease. Cold Spring Harb. Perspect. Med. 2014, 4, a015271. [Google Scholar] [CrossRef]
  43. Frank, A.P.; de Souza Santos, R.; Palmer, B.F.; Clegg, D.J. Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J. Lipid Res. 2019, 60, 1710–1719. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, M.J.; Fried, S.K. Sex-dependent Depot Differences in Adipose Tissue Development and Function; Role of Sex Steroids. J. Obes. Metab. Syndr. 2017, 26, 172–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jo, J.; Gavrilova, O.; Pack, S.; Jou, W.; Mullen, S.; Sumner, A.E.; Cushman, S.W.; Periwal, V. Hypertrophy and/or Hyperplasia: Dynamics of Adipose Tissue Growth. PLoS Comput. Biol. 2009, 5, e1000324. [Google Scholar] [CrossRef] [PubMed]
  46. Williams, R.; Pawlus, A.D.; Thornton, M.J. Getting under the skin of hair aging: The impact of the hair follicle environment. Exp. Dermatol. 2020, 29, 588–597. [Google Scholar] [CrossRef] [PubMed]
  47. Maares, M.; Haase, H. A Guide to Human Zinc Absorption: General Overview and Recent Advances of In Vitro Intestinal Models. Nutrients 2020, 12, 762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Hara, T.; Takeda, T.-A.; Takagishi, T.; Fukue, K.; Kambe, T.; Fukada, T. Physiological roles of zinc transporters: Molecular and genetic importance in zinc homeostasis. J. Physiol. Sci. 2017, 67, 283–301. [Google Scholar] [CrossRef]
  49. Ogawa, Y.; Kinoshita, M.; Shimada, S.; Kawamura, T. Zinc and Skin Disorders. Nutrients 2018, 10, 199. [Google Scholar] [CrossRef] [Green Version]
  50. Jackson, M. Physiology of zinc: General aspects. In Zinc in Human Biology; Springer: Berlin/Heidelberg, Germany, 1989; pp. 1–14. [Google Scholar]
  51. Michaelsson, G.; Ljunghall, K.; Danielson, B. Zinc in epidermis and dermis in healthy subjects. Acta Derm.-Venereol. 1980, 60, 295–299. [Google Scholar]
  52. Van Wouwe, J. Clinical and laboratory diagnosis of acrodermatitis enteropathica. Eur. J. Pediatr. 1989, 149, 2–8. [Google Scholar] [CrossRef]
  53. Wilson, D.; Varigos, G.; Ackland, M.L. Apoptosis may underlie the pathology of zinc-deficient skin. Immunol. Cell Biol. 2006, 84, 28–37. [Google Scholar] [CrossRef]
  54. Bao, B.; Prasad, A.S.; Beck, F.W.; Snell, D.; Suneja, A.; Sarkar, F.H.; Doshi, N.; Fitzgerald, J.T.; Swerdlow, P. Zinc supplementation decreases oxidative stress, incidence of infection, and generation of inflammatory cytokines in sickle cell disease patients. Transl. Res. 2008, 152, 67–80. [Google Scholar] [CrossRef] [PubMed]
  55. Xiang, Y.; Zhou, Q.; Li, Z.; Cui, Z.; Liu, X.; Liang, Y.; Zhu, S.; Zheng, Y.; Yeung, K.W.K.; Wu, S. A Z-scheme heterojunction of ZnO/CDots/C3N4 for strengthened photoresponsive bacteria-killing and acceleration of wound healing. J. Mater. Sci. Technol. 2020, 57, 1–11. [Google Scholar] [CrossRef]
  56. Pati, R.; Mehta, R.K.; Mohanty, S.; Padhi, A.; Sengupta, M.; Vaseeharan, B.; Goswami, C.; Sonawane, A. Topical application of zinc oxide nanoparticles reduces bacterial skin infection in mice and exhibits antibacterial activity by inducing oxidative stress response and cell membrane disintegration in macrophages. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 1195–1208. [Google Scholar] [CrossRef] [PubMed]
  57. Smijs, T.G.; Pavel, S. Titanium dioxide and zinc oxide nanoparticles in sunscreens: Focus on their safety and effectiveness. Nanotechnol. Sci. Appl. 2011, 4, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Pasquet, J.; Chevalier, Y.; Pelletier, J.; Couval, E.; Bouvier, D.; Bolzinger, M.-A. The contribution of zinc ions to the antimicrobial activity of zinc oxide. Colloids Surf. A Physicochem. Eng. Asp. 2014, 457, 263–274. [Google Scholar] [CrossRef]
  59. Pizzey, R.L.; Marquis, R.E.; Bradshaw, D.J. Antimicrobial effects of o-cymen-5-ol and zinc, alone & in combination in simple solutions and toothpaste formulations. Int. Dent. J. 2011, 61, 33–40. [Google Scholar]
  60. Sirelkhatim, A.; Mahmud, S.; Seeni, A.; Kaus, N.H.M.; Ann, L.C.; Bakhori, S.K.M.; Hasan, H.; Mohamad, D. Review on Zinc Oxide Nanoparticles: Antibacterial Activity and Toxicity Mechanism. Nano-Micro Lett. 2015, 7, 219–242. [Google Scholar] [CrossRef] [Green Version]
  61. Abebe, B.; Zereffa, E.A.; Tadesse, A.; Murthy, H. A review on enhancing the antibacterial activity of ZnO: Mechanisms and microscopic investigation. Nanoscale Res. Lett. 2020, 15, 190. [Google Scholar] [CrossRef]
  62. Chen, Y.; Cai, J.; Liu, D.; Liu, S.; Lei, D.; Zheng, L.; Wei, Q.; Gao, M. Zinc-based metal organic framework with antibacterial and anti-inflammatory properties for promoting wound healing. Regen. Biomater. 2022, 9, rbac019. [Google Scholar] [CrossRef]
  63. Liang, Y.; Liang, Y.; Zhang, H.; Guo, B. Antibacterial biomaterials for skin wound dressing. Asian J. Pharm. Sci. 2022, 17, 353–384. [Google Scholar] [CrossRef]
  64. Cortese-Krott, M.M.; Kulakov, L.; Opländer, C.; Kolb-Bachofen, V.; Kröncke, K.-D.; Suschek, C.V. Zinc regulates iNOS-derived nitric oxide formation in endothelial cells. Redox Biol. 2014, 2, 945–954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Yuan, S.; Sun, X.; Shen, Y.; Li, Z. Bioactive Poly (4-hydroxybutyrate)/Poly (ethylene glycol) Fibrous Dressings Incorporated with Zinc Oxide Nanoparticles for Efficient Antibacterial Therapy and Rapid Clotting. Macromol. Biosci. 2022, 22, 2100524. [Google Scholar] [CrossRef] [PubMed]
  66. Aslam, Z.; Roome, T.; Razzak, A.; Aslam, S.M.; Zaidi, M.B.; Kanwal, T.; Sikandar, B.; Bertino, M.F.; Rehman, K.; Shah, M.R. Investigation of wound healing potential of photo-active curcumin-ZnO-nanoconjugates in excisional wound model. Photodiagn. Photodyn. Ther. 2022, 39, 102956. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, H.; Ma, W.; Ma, H.; Qin, C.; Chen, J.; Wu, C. Spindle-Like Zinc Silicate Nanoparticles Accelerating Innervated and Vascularized Skin Burn Wound Healing. Adv. Healthc. Mater. 2022, 11, 2102359. [Google Scholar] [CrossRef] [PubMed]
  68. Hassan, A.; Sada, K.-K.; Ketheeswaran, S.; Dubey, A.K.; Bhat, M.S. Role of zinc in mucosal health and disease: A review of physiological, biochemical, and molecular processes. Cureus 2020, 12, e8197. [Google Scholar] [CrossRef]
  69. Eide, D.J. Zinc transporters and the cellular trafficking of zinc. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2006, 1763, 711–722. [Google Scholar] [CrossRef]
  70. Jeong, J.; Eide, D.J. The SLC39 family of zinc transporters. Mol. Asp. Med. 2013, 34, 612–619. [Google Scholar] [CrossRef] [Green Version]
  71. Styrpejko, D.J.; Cuajungco, M.P. Transmembrane 163 (TMEM163) Protein: A New Member of the Zinc Efflux Transporter Family. Biomedicines 2021, 9, 220. [Google Scholar] [CrossRef]
  72. Hoch, E.; Levy, M.; Hershfinkel, M.; Sekler, I. Elucidating the H+ Coupled Zn2+ Transport Mechanism of ZIP4; Implications in Acrodermatitis Enteropathica. Int. J. Mol. Sci. 2020, 21, 734. [Google Scholar] [CrossRef] [Green Version]
  73. Bin, B.H.; Seo, J.; Kim, S.T. Function, Structure, and Transport Aspects of ZIP and ZnT Zinc Transporters in Immune Cells. J. Immunol. Res. 2018, 2018, 9365747. [Google Scholar] [CrossRef] [Green Version]
  74. Liu, Z.; Li, H.; Soleimani, M.; Girijashanker, K.; Reed, J.M.; He, L.; Dalton, T.P.; Nebert, D.W. Cd2+ versus Zn2+ uptake by the ZIP8 HCO3-dependent symporter: Kinetics, electrogenicity and trafficking. Biochem. Biophys. Res. Commun. 2008, 365, 814–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lin, W.; Chai, J.; Love, J.; Fu, D. Selective electrodiffusion of zinc ions in a Zrt-, Irt-like protein, ZIPB. J. Biol. Chem. 2010, 285, 39013–39020. [Google Scholar] [CrossRef] [Green Version]
  76. Bin, B.H.; Hojyo, S.; Seo, J.; Hara, T.; Takagishi, T.; Mishima, K.; Fukada, T. The Role of the Slc39a Family of Zinc Transporters in Zinc Homeostasis in Skin. Nutrients 2018, 10, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Bin, B.-H.; Bhin, J.; Takaishi, M.; Toyoshima, K.-E.; Kawamata, S.; Ito, K.; Hara, T.; Watanabe, T.; Irié, T.; Takagishi, T.; et al. Requirement of zinc transporter ZIP10 for epidermal development: Implication of the ZIP10–p63 axis in epithelial homeostasis. Proc. Natl. Acad. Sci. USA 2017, 114, 12243–12248. [Google Scholar] [PubMed] [Green Version]
  78. Lažetić, V.; Wu, F.; Cohen, L.B.; Reddy, K.C.; Chang, Y.-T.; Gang, S.S.; Bhabha, G.; Troemel, E.R. The transcription factor ZIP-1 promotes resistance to intracellular infection in Caenorhabditis elegans. Nat. Commun. 2022, 13, 17. [Google Scholar] [CrossRef] [PubMed]
  79. Bin, B.H.; Lee, S.H.; Bhin, J.; Irié, T.; Kim, S.; Seo, J.; Mishima, K.; Lee, T.R.; Hwang, D.; Fukada, T.; et al. The epithelial zinc transporter ZIP10 epigenetically regulates human epidermal homeostasis by modulating histone acetyltransferase activity. Br. J. Dermatol. 2019, 180, 869–880. [Google Scholar] [CrossRef] [PubMed]
  80. Lee, M.G.; Bin, B.H. Different Actions of Intracellular Zinc Transporters ZIP7 and ZIP13 Are Essential for Dermal Development. Int. J. Mol. Sci. 2019, 20, 3941. [Google Scholar] [CrossRef] [Green Version]
  81. Subramanian Vignesh, K.; Deepe, G.S., Jr. Metallothioneins: Emerging Modulators in Immunity and Infection. Int. J. Mol. Sci. 2017, 18, 2197. [Google Scholar] [CrossRef] [Green Version]
  82. Jagadeesan, S.; Kaliyadan, F. Acrodermatitis Enteropathica; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  83. Bin, B.H.; Hojyo, S.; Ryong Lee, T.; Fukada, T. Spondylocheirodysplastic Ehlers-Danlos syndrome (SCD-EDS) and the mutant zinc transporter ZIP13. Rare Dis. 2014, 2, e974982. [Google Scholar] [CrossRef] [Green Version]
  84. Ramoz, N.; Rueda, L.A.; Bouadjar, B.; Montoya, L.S.; Orth, G.; Favre, M. Mutations in two adjacent novel genes are associated with epidermodysplasia verruciformis. Nat. Genet. 2002, 32, 579–581. [Google Scholar] [CrossRef]
  85. Ramoz, N.; Taïeb, A.; Rueda, L.A.; Montoya, L.S.; Bouadjar, B.; Favre, M.; Orth, G. Evidence for a nonallelic heterogeneity of epidermodysplasia verruciformis with two susceptibility loci mapped to chromosome regions 2p21-p24 and 17q25. J. Investig. Dermatol. 2000, 114, 1148–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Orth, G. Host defenses against human papillomaviruses: Lessons from epidermodysplasia verruciformis. Curr. Top. Microbiol. Immunol. 2008, 321, 59–83. [Google Scholar] [PubMed]
  87. Golan, Y.; Kambe, T.; Assaraf, Y.G. The role of the zinc transporter SLC30A2/ZnT2 in transient neonatal zinc deficiency. Met. Integr. Biometal Sci. 2017, 9, 1352–1366. [Google Scholar] [CrossRef] [PubMed]
  88. Miletta, M.C.; Bieri, A.; Kernland, K.; Schöni, M.H.; Petkovic, V.; Flück, C.E.; Eblé, A.; Mullis, P.E. Transient Neonatal Zinc Deficiency Caused by a Heterozygous G87R Mutation in the Zinc Transporter ZnT-2 (SLC30A2) Gene in the Mother Highlighting the Importance of Zn2+ for Normal Growth and Development. Int. J. Endocrinol. 2013, 2013, 259189. [Google Scholar] [CrossRef] [Green Version]
  89. Gammoh, N.Z.; Rink, L. Zinc in Infection and Inflammation. Nutrients 2017, 9, 624. [Google Scholar] [CrossRef] [Green Version]
  90. Kambe, T.; Yamaguchi-Iwai, Y.; Sasaki, R.; Nagao, M. Overview of mammalian zinc transporters. Cell. Mol. Life Sci. CMLS 2004, 61, 49–68. [Google Scholar] [CrossRef]
  91. Huang, L.; Kirschke, C.P.; Zhang, Y.; Yu, Y.Y. The ZIP7 gene (Slc39a7) encodes a zinc transporter involved in zinc homeostasis of the Golgi apparatus. J. Biol. Chem. 2005, 280, 15456–15463. [Google Scholar] [CrossRef] [Green Version]
  92. Bin, B.-H.; Bhin, J.; Seo, J.; Kim, S.-Y.; Lee, E.; Park, K.; Choi, D.-H.; Takagishi, T.; Hara, T.; Hwang, D.; et al. Requirement of Zinc Transporter SLC39A7/ZIP7 for Dermal Development to Fine-Tune Endoplasmic Reticulum Function by Regulating Protein Disulfide Isomerase. J. Investig. Dermatol. 2017, 137, 1682–1691. [Google Scholar] [CrossRef]
  93. Fukada, T.; Civic, N.; Furuichi, T.; Shimoda, S.; Mishima, K.; Higashiyama, H.; Idaira, Y.; Asada, Y.; Kitamura, H.; Yamasaki, S.; et al. The zinc transporter SLC39A13/ZIP13 is required for connective tissue development; its involvement in BMP/TGF-beta signaling pathways. PLoS ONE 2008, 3, e3642. [Google Scholar] [CrossRef]
  94. Jeong, J.; Walker, J.M.; Wang, F.; Park, J.G.; Palmer, A.E.; Giunta, C.; Rohrbach, M.; Steinmann, B.; Eide, D.J. Promotion of vesicular zinc efflux by ZIP13 and its implications for spondylocheiro dysplastic Ehlers–Danlos syndrome. Proc. Natl. Acad. Sci. USA 2012, 109, E3530–E3538. [Google Scholar] [CrossRef] [Green Version]
  95. Bin, B.H.; Fukada, T.; Hosaka, T.; Yamasaki, S.; Ohashi, W.; Hojyo, S.; Miyai, T.; Nishida, K.; Yokoyama, S.; Hirano, T. Biochemical characterization of human ZIP13 protein: A homo-dimerized zinc transporter involved in the spondylocheiro dysplastic Ehlers-Danlos syndrome. J. Biol. Chem. 2011, 286, 40255–40265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Adulcikas, J.; Norouzi, S.; Bretag, L.; Sohal, S.S.; Myers, S. The zinc transporter SLC39A7 (ZIP7) harbours a highly-conserved histidine-rich N-terminal region that potentially contributes to zinc homeostasis in the endoplasmic reticulum. Comput. Biol. Med. 2018, 100, 196–202. [Google Scholar] [CrossRef] [PubMed]
  97. Anzilotti, C.; Swan, D.J.; Boisson, B.; Deobagkar-Lele, M.; Oliveira, C.; Chabosseau, P.; Engelhardt, K.R.; Xu, X.; Chen, R.; Alvarez, L.; et al. An essential role for the Zn2+ transporter ZIP7 in B cell development. Nat. Immunol. 2019, 20, 350–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Xu, J.; Wan, Z.; Zhou, B. Drosophila ZIP13 is posttranslationally regulated by iron-mediated stabilization. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2019, 1866, 1487–1497. [Google Scholar] [CrossRef] [PubMed]
  99. Guo, L.; Li, X.; Tang, Q.-Q. Transcriptional regulation of adipocyte differentiation: A central role for CCAAT/enhancer-binding protein (C/EBP) β. J. Biol. Chem. 2015, 290, 755–761. [Google Scholar] [CrossRef] [Green Version]
  100. Fukunaka, A.; Fukada, T.; Bhin, J.; Suzuki, L.; Tsuzuki, T.; Takamine, Y.; Bin, B.-H.; Yoshihara, T.; Ichinoseki-Sekine, N.; Naito, H. Zinc transporter ZIP13 suppresses beige adipocyte biogenesis and energy expenditure by regulating C/EBP-β expression. PLoS Genet. 2017, 13, e1006950. [Google Scholar] [CrossRef]
  101. Skalny, A.V.; Aschner, M.; Tinkov, A.A. Zinc. Adv. Food Nutr. Res. 2021, 96, 251–310. [Google Scholar]
  102. Narváez-Caicedo, C.; Moreano, G.; Sandoval, B.A.; Jara-Palacios, M. Zinc Deficiency among Lactating Mothers from a Peri-Urban Community of the Ecuadorian Andean Region: An Initial Approach to the Need of Zinc Supplementation. Nutrients 2018, 10, 869. [Google Scholar] [CrossRef] [Green Version]
  103. Rokunohe, D.; Nakano, H.; Ikenaga, S.; Umegaki, N.; Kaneko, T.; Matsuhashi, Y.; Tando, Y.; Toyoki, Y.; Hakamada, K.; Kusumi, T. Reduction in epidermal Langerhans cells in patients with necrolytic migratory erythema. J. Dermatol. Sci. 2008, 50, 76–80. [Google Scholar] [CrossRef]
  104. Tierney, E.P.; Badger, J. Etiology and pathogenesis of necrolytic migratory erythema: Review of the literature. Medscape Gen. Med. 2004, 6, 4. [Google Scholar]
  105. Vannucchi, H.; Fávaro, R.M.; Cunha, D.F.; Marchini, J.S. Assessment of zinc nutritional status of pellagra patients. Alcohol Alcohol. 1995, 30, 297–302. [Google Scholar] [PubMed]
  106. Nakamura, H.; Sekiguchi, A.; Ogawa, Y.; Kawamura, T.; Akai, R.; Iwawaki, T.; Makiguchi, T.; Yokoo, S.; Ishikawa, O.; Motegi, S.I. Zinc deficiency exacerbates pressure ulcers by increasing oxidative stress and ATP in the skin. J. Dermatol. Sci. 2019, 95, 62–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Krebs, N.F.; Miller, L.V.; Hambidge, K.M. Zinc deficiency in infants and children: A review of its complex and synergistic interactions. Paediatr. Int. Child Health 2014, 34, 279–288. [Google Scholar] [CrossRef] [PubMed]
  108. Schmitt, S.; Küry, S.; Giraud, M.; Dréno, B.; Kharfi, M.; Bézieau, S. An update on mutations of the SLC39A4 gene in acrodermatitis enteropathica. Hum. Mutat. 2009, 30, 926–933. [Google Scholar] [CrossRef] [PubMed]
  109. Maverakis, E.; Fung, M.A.; Lynch, P.J.; Draznin, M.; Michael, D.J.; Ruben, B.; Fazel, N. Acrodermatitis enteropathica and an overview of zinc metabolism. J. Am. Acad. Dermatol. 2007, 56, 116–124. [Google Scholar] [CrossRef] [PubMed]
  110. Perafán-Riveros, C.; França, L.F.S.; Alves, A.C.F.; Sanches, J.A., Jr. Acrodermatitis enteropathica: Case report and review of the literature. Pediatr. Dermatol. 2002, 19, 426–431. [Google Scholar] [CrossRef] [PubMed]
  111. Nistor, N.; Ciontu, L.; Frasinariu, O.-E.; Lupu, V.V.; Ignat, A.; Streanga, V. Acrodermatitis enteropathica: A case report. Medicine 2016, 95, e3553. [Google Scholar] [CrossRef] [PubMed]
  112. Krieger, I.; Evans, G. Acrodermatitis enteropathica without hypozincemia: Therapeutic effect of a pancreatic enzyme preparation due to a zinc-binding ligand. J. Pediatr. 1980, 96, 32–35. [Google Scholar] [CrossRef]
  113. Kim, Y.J.; Kim, M.Y.; Kim, H.O.; Lee, M.D.; Park, Y.M. Acrodermatitis enteropathica-like eruption associated with combined nutritional deficiency. J. Korean Med. Sci. 2005, 20, 908–911. [Google Scholar] [CrossRef]
  114. Jensen, S.L.; McCuaig, C.; Zembowicz, A.; Hurt, M.A. Bullous lesions in acrodermatitis enteropathica delaying diagnosis of zinc deficiency: A report of two cases and review of the literature. J. Cutan. Pathol. 2008, 35, 1–13. [Google Scholar] [CrossRef]
  115. Zimmerman, A.W.; Hambidge, K.M.; Lepow, M.L.; Greenberg, R.D.; Stover, M.L.; Casey, C.E. Acrodermatitis in breast-fed premature infants: Evidence for a defect of mammary zinc secretion. Pediatrics 1982, 69, 176–183. [Google Scholar] [CrossRef] [PubMed]
  116. Sehgal, V.N.; Jain, S. Acrodermatitis enteropathica. Clin. Dermatol. 2000, 18, 745–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Takeda, T.-a.; Miyazaki, S.; Kobayashi, M.; Nishino, K.; Goto, T.; Matsunaga, M.; Ooi, M.; Shirakawa, H.; Tani, F.; Kawamura, T. Zinc deficiency causes delayed ATP clearance and adenosine generation in rats and cell culture models. Commun. Biol. 2018, 1, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. al-Rashida, M.; Iqbal, J. Therapeutic Potentials of Ecto-Nucleoside Triphosphate Diphosphohydrolase, Ecto-Nucleotide Pyrophosphatase/Phosphodiesterase, Ecto-5′-Nucleotidase, and Alkaline Phosphatase Inhibitors. Med. Res. Rev. 2014, 34, 703–743. [Google Scholar] [CrossRef]
  119. Fujimoto, S.; Tsuji, T.; Fujiwara, T.; Takeda, T.-a.; Merriman, C.; Fukunaka, A.; Nishito, Y.; Fu, D.; Hoch, E.; Sekler, I. The PP-motif in luminal loop 2 of ZnT transporters plays a pivotal role in TNAP activation. Biochem. J. 2016, 473, 2611–2621. [Google Scholar] [CrossRef] [Green Version]
  120. Kawamura, T.; Ogawa, Y.; Nakamura, Y.; Nakamizo, S.; Ohta, Y.; Nakano, H.; Kabashima, K.; Katayama, I.; Koizumi, S.; Kodama, T.; et al. Severe dermatitis with loss of epidermal Langerhans cells in human and mouse zinc deficiency. J. Clin. Investig. 2012, 122, 722–732. [Google Scholar] [CrossRef] [Green Version]
  121. Ogawa, Y.; Kinoshita, M.; Shimada, S.; Kawamura, T. Zinc in Keratinocytes and Langerhans Cells: Relevance to the Epidermal Homeostasis. J. Immunol. Res. 2018, 2018, 5404093. [Google Scholar] [CrossRef] [Green Version]
  122. Mizumoto, N.; Kumamoto, T.; Robson, S.C.; Sévigny, J.; Matsue, H.; Enjyoji, K.; Takashima, A. CD39 is the dominant Langerhans cell–associated ecto-NTPDase: Modulatory roles in inflammation and immune responsiveness. Nat. Med. 2002, 8, 358–365. [Google Scholar] [CrossRef]
  123. Ogawa, Y.; Kinoshita, M.; Mizumura, N.; Miyazaki, S.; Aoki, R.; Momosawa, A.; Shimada, S.; Kambe, T.; Kawamura, T. Purinergic Molecules in the Epidermis. J. Investig. Dermatol. 2018, 138, 2486–2488. [Google Scholar] [CrossRef] [Green Version]
  124. Castori, M. Ehlers-danlos syndrome, hypermobility type: An underdiagnosed hereditary connective tissue disorder with mucocutaneous, articular, and systemic manifestations. ISRN Dermatol. 2012, 2012, 751768. [Google Scholar] [CrossRef] [Green Version]
  125. Giunta, C.; Elçioglu, N.H.; Albrecht, B.; Eich, G.; Chambaz, C.; Janecke, A.R.; Yeowell, H.; Weis, M.; Eyre, D.R.; Kraenzlin, M.; et al. Spondylocheiro dysplastic form of the Ehlers-Danlos syndrome--an autosomal-recessive entity caused by mutations in the zinc transporter gene SLC39A13. Am. J. Hum. Genet. 2008, 82, 1290–1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Guo, X.; Wang, X.F. Signaling cross-talk between TGF-beta/BMP and other pathways. Cell Res. 2009, 19, 71–88. [Google Scholar] [CrossRef] [PubMed]
  127. Abreu, J.G.; Ketpura, N.I.; Reversade, B.; De Robertis, E. Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-β. Nat. Cell Biol. 2002, 4, 599–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Kim, D.; Kim, S.H.; Li, G.C. Proteasome inhibitors MG132 and lactacystin hyperphosphorylate HSF1 and induce hsp70 and hsp27 expression. Biochem. Biophys. Res. Commun. 1999, 254, 264–268. [Google Scholar] [CrossRef]
  129. Huang, L.; Chen, C.H. Proteasome regulators: Activators and inhibitors. Curr. Med. Chem. 2009, 16, 931–939. [Google Scholar] [CrossRef] [Green Version]
  130. Tanaka, K. The proteasome: Overview of structure and functions. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 12–36. [Google Scholar] [CrossRef] [Green Version]
  131. Jang, H.H. Regulation of Protein Degradation by Proteasomes in Cancer. J. Cancer Prev. 2018, 23, 153–161. [Google Scholar] [CrossRef]
  132. Cohen-Kaplan, V.; Livneh, I.; Avni, N.; Fabre, B.; Ziv, T.; Kwon, Y.T.; Ciechanover, A. p62-and ubiquitin-dependent stress-induced autophagy of the mammalian 26S proteasome. Proc. Natl. Acad. Sci. USA 2016, 113, E7490–E7499. [Google Scholar] [CrossRef]
  133. Besche, H.C.; Sha, Z.; Kukushkin, N.V.; Peth, A.; Hock, E.M.; Kim, W.; Gygi, S.; Gutierrez, J.A.; Liao, H.; Dick, L. Autoubiquitination of the 26S proteasome on Rpn13 regulates breakdown of ubiquitin conjugates. EMBO J. 2014, 33, 1159–1176. [Google Scholar] [CrossRef] [Green Version]
  134. Pancheri, E.; Guglielmi, V.; Wilczynski, G.M.; Malatesta, M.; Tonin, P.; Tomelleri, G.; Nowis, D.; Vattemi, G. Non-Hematologic Toxicity of Bortezomib in Multiple Myeloma: The Neuromuscular and Cardiovascular Adverse Effects. Cancers 2020, 12, 2540. [Google Scholar] [CrossRef]
  135. Starheim, K.K.; Holien, T.; Misund, K.; Johansson, I.; Baranowska, K.A.; Sponaas, A.M.; Hella, H.; Buene, G.; Waage, A.; Sundan, A. Intracellular glutathione determines bortezomib cytotoxicity in multiple myeloma cells. Blood Cancer J. 2016, 6, e446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shiber, A.; Ravid, T. Chaperoning proteins for destruction: Diverse roles of Hsp70 chaperones and their co-chaperones in targeting misfolded proteins to the proteasome. Biomolecules 2014, 4, 704–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Esser, C.; Alberti, S.; Höhfeld, J. Cooperation of molecular chaperones with the ubiquitin/proteasome system. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2004, 1695, 171–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Bin, B.H.; Hojyo, S.; Hosaka, T.; Bhin, J.; Kano, H.; Miyai, T.; Ikeda, M.; Kimura-Someya, T.; Shirouzu, M.; Cho, E.G.; et al. Molecular pathogenesis of spondylocheirodysplastic Ehlers-Danlos syndrome caused by mutant ZIP13 proteins. EMBO Mol. Med. 2014, 6, 1028–1042. [Google Scholar] [CrossRef] [PubMed]
  139. Kogan, S.; Sood, A.; Garnick, M. Zinc and Wound Healing: A Review of Zinc Physiology and Clinical Applications. Wounds A Compend. Clin. Res. Pract. 2017, 29, 102–106. [Google Scholar]
  140. Toyoda, M.; Morohashi, M. Pathogenesis of acne. Med. Electron Microsc. 2001, 34, 29–40. [Google Scholar] [CrossRef] [PubMed]
  141. Cong, T.-X.; Hao, D.; Wen, X.; Li, X.-H.; He, G.; Jiang, X. From pathogenesis of acne vulgaris to anti-acne agents. Arch. Dermatol. Res. 2019, 311, 337–349. [Google Scholar] [CrossRef]
  142. Beylot, C.; Auffret, N.; Poli, F.; Claudel, J.-P.; Leccia, M.-T.; Del Giudice, P.; Dreno, B. Propionibacterium acnes: An update on its role in the pathogenesis of acne. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 271–278. [Google Scholar] [CrossRef]
  143. Gollnick, H. Current Concepts of the Pathogenesis of Acne. Drugs 2003, 63, 1579–1596. [Google Scholar] [CrossRef]
  144. Dreno, B.; Gollnick, H.P.M.; Kang, S.; Thiboutot, D.; Bettoli, V.; Torres, V.; Leyden, J.; the Global Alliance to Improve Outcomes in Acne. Understanding innate immunity and inflammation in acne: Implications for management. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 3–11. [Google Scholar] [CrossRef] [Green Version]
  145. Cervantes, J.; Eber, A.E.; Perper, M.; Nascimento, V.M.; Nouri, K.; Keri, J.E. The role of zinc in the treatment of acne: A review of the literature. Dermatol. Ther. 2018, 31, e12576. [Google Scholar] [CrossRef] [PubMed]
  146. Yee, B.E.; Richards, P.; Sui, J.Y.; Marsch, A.F. Serum zinc levels and efficacy of zinc treatment in acne vulgaris: A systematic review and meta-analysis. Dermatol. Ther. 2020, 33, e14252. [Google Scholar] [CrossRef] [PubMed]
  147. Lidén, S.; Göransson, K.; Odsell, L. Clinical evaluation in acne. Acta Derm.-Venereol. Suppl. 1980, 89 (Suppl. 89), 47–52. [Google Scholar]
  148. Gupta, M.; Mahajan, V.K.; Mehta, K.S.; Chauhan, P.S. Zinc therapy in dermatology: A review. Dermatol. Res. Pract. 2014, 2014, 709152. [Google Scholar] [CrossRef] [PubMed]
  149. Papageorgiou, P.; Chu, C. Chloroxylenol and zinc oxide containing cream (Nels cream®) vs. 5% benzoyl peroxide cream in the treatment of acne vulgaris. A double-blind, randomized, controlled trial. Clin. Exp. Dermatol. 2000, 25, 16–20. [Google Scholar] [CrossRef]
  150. Michaëlsson, G.; Juhlin, L.; Vahlquist, A. Effects of Oral Zinc and Vitamin A in Acne. Arch. Dermatol. 1977, 113, 31–36. [Google Scholar] [CrossRef]
  151. Vahlquist, A.; Michaëlsson, G.; Juhlin, L. Acne treatment with oral zinc and vitamin A: Effects on the serum levels of zinc and retinol binding protein (RBP). Acta Derm.-Venereol. 1978, 58, 437–442. [Google Scholar]
  152. Kobayashi, H.; Aiba, S.; Tagami, H. Successful treatment of dissecting cellulitis and acne conglobata with oral zinc. Br. J. Dermatol. 1999, 141, 1136–1152. [Google Scholar] [CrossRef]
  153. Weimar, V.M.; Puhl, S.C.; Smith, W.H.; tenBroeke, J.E. Zinc Sulfate in Acne Vulgaris. Arch. Dermatol. 1978, 114, 1776–1778. [Google Scholar] [CrossRef]
  154. Nast, A.; Dreno, B.; Bettoli, V.; Degitz, K.; Erdmann, R.; Finlay, A.; Ganceviciene, R.; Haedersdal, M.; Layton, A.; López-Estebaranz, J. European evidence-based (S3) guidelines for the treatment of acne. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 1–29. [Google Scholar] [CrossRef]
  155. Hillstrom, L.; Pettersson, L.; Hellbe, L.; Kjellin, A.; Leczinsky, C.G.; Nordwall, C. Comparison of oral treatment with zinc sulphate and placebo in acne vulgaris. Br. J. Dermatol. 1977, 97, 679–684. [Google Scholar] [CrossRef]
  156. Verma, K.; Saini, A.; Dhamija, S. Oral zinc sulphate therapy in acne vulgaris: A double-blind trial. Acta Derm.-Venereol. 1980, 60, 337–340. [Google Scholar] [PubMed]
  157. Dreno, B.; Moyse, D.; Alirezai, M.; Amblard, P.; Auffret, N.; Beylot, C.; Bodokh, I.; Chivot, M.; Daniel, F.; Humbert, P. Multicenter randomized comparative double-blind controlled clinical trial of the safety and efficacy of zinc gluconate versus minocycline hydrochloride in the treatment of inflammatory acne vulgaris. Dermatology 2001, 203, 135–140. [Google Scholar] [CrossRef] [PubMed]
  158. Katsambas, A.; Dessinioti, C. New and emerging treatments in dermatology: Acne. Dermatol. Ther. 2008, 21, 86–95. [Google Scholar] [CrossRef] [PubMed]
  159. Bowe, W.P.; Joshi, S.S.; Shalita, A.R. Diet and acne. J. Am. Acad. Dermatol. 2010, 63, 124–141. [Google Scholar] [CrossRef]
  160. Sardana, K.; Garg, V.K. An observational study of methionine-bound zinc with antioxidants for mild to moderate acne vulgaris. Dermatol. Ther. 2010, 23, 411–418. [Google Scholar] [CrossRef]
  161. Walocko, F.M.; Eber, A.E.; Keri, J.E.; Al-harbi, M.A.; Nouri, K. The role of nicotinamide in acne treatment. Dermatol. Ther. 2017, 30, e12481. [Google Scholar] [CrossRef]
  162. Niren, N.M.; Torok, H.M. The Nicomide Improvement in Clinical Outcomes Study (NICOS): Results of an 8-week trial. Cutis 2006, 77 (Suppl. 1), 17–28. [Google Scholar]
  163. Sharquie, K.E.; Al-Mashhadani, S.A.; Noaimi, A.A.; Hasan, A.A. Topical zinc sulphate (25%) solution: A new therapy for actinic keratosis. J. Cutan. Aesthetic Surg. 2012, 5, 53–56. [Google Scholar] [CrossRef]
  164. Park, H.; Kim, C.W.; Kim, S.S.; Park, C.W. The Therapeutic Effect and the Changed Serum Zinc Level after Zinc Supplementation in Alopecia Areata Patients Who Had a Low Serum Zinc Level. Ann. Dermatol. 2009, 21, 142–146. [Google Scholar] [CrossRef] [Green Version]
  165. Lux-Battistelli, C. Combination therapy with zinc gluconate and PUVA for alopecia areata totalis: An adjunctive but crucial role of zinc supplementation. Dermatol. Ther. 2015, 28, 235–238. [Google Scholar] [CrossRef] [PubMed]
  166. Schwartz, J.R.; Marsh, R.G.; Draelos, Z.D. Zinc and Skin Health: Overview of Physiology and Pharmacology. Dermatol. Surg. 2005, 31, 837–847. [Google Scholar] [CrossRef] [PubMed]
  167. Chretien, J.H.; Esswein, J.G.; Sharpe, L.M.; Kiely, J.J.; Lyddon, F.E. Efficacy of Undecylenic Acid–Zinc Undecylenate Powder in Culture Positive Tinea Pedis. Int. J. Dermatol. 1980, 19, 51–54. [Google Scholar] [CrossRef] [PubMed]
  168. Famenini, S.; Goh, C. Evidence for supplemental treatments in androgenetic alopecia. J. Drugs Dermatol. 2014, 13, 809–812. [Google Scholar] [PubMed]
  169. Berger, R.S.; Fu, J.L.; Smiles, K.A.; Turner, C.B.; Schnell, B.M.; Werchowski, K.M.; Lammers, K.M. The effects of minoxidil, 1% pyrithione zinc and a combination of both on hair density: A randomized controlled trial. Br. J. Dermatol. 2003, 149, 354–362. [Google Scholar] [CrossRef]
  170. Sharquie, K.E.; Najim, R.A.; Al-Dori, W.S.; Al-Hayani, R.K. Oral zinc sulfate in the treatment of Behcet’s disease: A double blind cross-over study. J. Dermatol. 2006, 33, 541–546. [Google Scholar] [CrossRef]
  171. Bulur, I.; Onder, M. Behçet disease: New aspects. Clin. Dermatol. 2017, 35, 421–434. [Google Scholar] [CrossRef]
  172. Ascione, J.-M.; Forestier, S.; Rollat-Corvol, I. Deodorant Composition Comprising a Water-Soluble Zinc Salt as Odor-Absorbing Agent. U.S. Patent 6,632,421, 14 October 2003. [Google Scholar]
  173. Li, W.; Liu, M. Method and Composition for Preventing Sweat-Related Odor. U.S. Patent 6,426,061, 30 July 2002. [Google Scholar]
  174. Sharquie, K.E.; Noaimi, A.A.; Hameed, S.D. Topical 15% zinc sulfate solution is an effective therapy for feet odor. J. Cosmet. Dermatol. Sci. Appl. 2013, 3, 35867. [Google Scholar] [CrossRef] [Green Version]
  175. Sharquie, K.E.; Najim, R.A.; Farjou, I.B. A comparative controlled trial of intralesionally-administered zinc sulphate, hypertonic sodium chloride and pentavalent antimony compound against acute cutaneous leishmaniasis. Clin. Exp. Dermatol. 1997, 22, 169–173. [Google Scholar] [CrossRef]
  176. Sharquie, K.E.; Najim, R.A.; Farjou, I.B.; Al-Timimi, D.J. Oral zinc sulphate in the treatment of acute cutaneous leishmaniasis. Clin. Exp. Dermatol. 2001, 26, 21–26. [Google Scholar] [CrossRef]
  177. Minodier, P.; Parola, P. Cutaneous leishmaniasis treatment. Travel Med. Infect. Dis. 2007, 5, 150–158. [Google Scholar] [CrossRef] [PubMed]
  178. Berne, B.; Venge, P.; Öhman, S. Perifolliculitis Capitis Abscedens et Suffodiens (Hoffman): Complete Healing Associated With Oral Zinc Therapy. Arch. Dermatol. 1985, 121, 1028–1030. [Google Scholar] [CrossRef] [PubMed]
  179. Simpson, N.B.; Cunliffe, W.J. Disorders of the Sebaceous Glands. In Rook’s Textbook of Dermatology; Blackwell Science Ltd.: Hoboken, NJ, USA, 2004; pp. 2121–2196. [Google Scholar]
  180. Wiegand, C.; Hipler, U.C.; Boldt, S.; Strehle, J.; Wollina, U. Skin-protective effects of a zinc oxide-functionalized textile and its relevance for atopic dermatitis. Clin. Cosmet. Investig. Dermatol. 2013, 6, 115–121. [Google Scholar] [PubMed] [Green Version]
  181. Baldwin, S.; Odio, M.R.; Haines, S.L.; O’Connor, R.J.; Englehart, J.S.; Lane, A.T. Skin benefits from continuous topical administration of a zinc oxide/petrolatum formulation by a novel disposable diaper. J. Eur. Acad. Dermatol. Venereol. 2001, 15, 5–11. [Google Scholar] [CrossRef] [PubMed]
  182. Faghihi, G.; Iraji, F.; Shahingohar, A.; Saidat, A. The efficacy of ‘0.05% Clobetasol + 2.5% zinc sulphate’ cream vs. ‘0.05% Clobetasol alone’ cream in the treatment of the chronic hand eczema: A double-blind study. J. Eur. Acad. Dermatol. Venereol. 2008, 22, 531–536. [Google Scholar] [CrossRef] [PubMed]
  183. Ikeda, M.; Arata, J.; Isaka, H. Erosive pustular dermatosis of the scalp successfully treated with oral zinc sulphate. Br. J. Dermatol. 1982, 106, 742. [Google Scholar] [CrossRef]
  184. Fernández-Romero, J.A.; Abraham, C.J.; Rodriguez, A.; Kizima, L.; Jean-Pierre, N.; Menon, R.; Begay, O.; Seidor, S.; Ford, B.E.; Gil, P.I. Zinc acetate/carrageenan gels exhibit potent activity in vivo against high-dose herpes simplex virus 2 vaginal and rectal challenge. Antimicrob. Agents Chemother. 2012, 56, 358–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Kenney, J.; Rodríguez, A.; Kizima, L.; Seidor, S.; Menon, R.; Jean-Pierre, N.; Pugach, P.; Levendosky, K.; Derby, N.; Gettie, A.; et al. A Modified Zinc Acetate Gel, a Potential Nonantiretroviral Microbicide, Is Safe and Effective against Simian-Human Immunodeficiency Virus and Herpes Simplex Virus 2 Infection In Vivo. Antimicrob. Agents Chemother. 2013, 57, 4001–4009. [Google Scholar] [CrossRef] [Green Version]
  186. Tavakoli, A.; Ataei-Pirkooh, A.; Mm Sadeghi, G.; Bokharaei-Salim, F.; Sahrapour, P.; Kiani, S.J.; Moghoofei, M.; Farahmand, M.; Javanmard, D.; Monavari, S.H. Polyethylene glycol-coated zinc oxide nanoparticle: An efficient nanoweapon to fight against herpes simplex virus type 1. Nanomedicine 2018, 13, 2675–2690. [Google Scholar] [CrossRef]
  187. Arens, M.; Travis, S. Zinc Salts Inactivate Clinical Isolates of Herpes Simplex Virus In Vitro. J. Clin. Microbiol. 2000, 38, 1758–1762. [Google Scholar] [CrossRef] [Green Version]
  188. Brocard, A.; Knol, A.C.; Khammari, A.; Dréno, B. Hidradenitis Suppurativa and Zinc: A New Therapeutic Approach. Dermatology 2007, 214, 325–327. [Google Scholar] [CrossRef] [PubMed]
  189. Hessam, S.; Sand, M.; Meier, N.M.; Gambichler, T.; Scholl, L.; Bechara, F.G. Combination of oral zinc gluconate and topical triclosan: An anti-inflammatory treatment modality for initial hidradenitis suppurativa. J. Dermatol. Sci. 2016, 84, 197–202. [Google Scholar] [CrossRef] [PubMed]
  190. Battistini, F.; Cordero, C.; Urcuyo, F.G.; Rojas, R.F.; Ollague, W.; Zaias, N. The Treatment of Dermatophytoses of the Glabrous Skin: A Comparison of Undecylenic Acid and its Salt Versus Tolnaftate. Int. J. Dermatol. 1983, 22, 388–389. [Google Scholar] [CrossRef] [PubMed]
  191. Mathur, N.K.; Bumb, R.A.; Mangal, H.N.; Sharma, M.L. Oral zinc as an adjunct to dapsone in lepromatous leprosy. Int. J. Lepr. 1984, 52, 331–338. [Google Scholar]
  192. Sehgal, V.N.; Prasad, P.V.S.; Kaviarasan, P.K.; Rajan, D. Trophic skin ulceration in leprosy: Evaluation of the efficacy of topical phenytoin sodium zinc oxide paste. Int. J. Dermatol. 2014, 53, 873–878. [Google Scholar] [CrossRef] [PubMed]
  193. Cuevas, L.E.; Koyanagi, A. Zinc and infection: A review. Ann. Trop. Paediatr. 2005, 25, 149–160. [Google Scholar] [CrossRef]
  194. Sharquie, K.E.; Al-Mashhadani, S.A.; Salman, H.A. Topical 10% Zinc Sulfate Solution for Treatment of Melasma. Dermatol. Surg. 2008, 34, 1346–1349. [Google Scholar]
  195. Castanedo-Cazares, J.P.; Hernandez-Blanco, D.; Carlos-Ortega, B.; Fuentes-Ahumada, C.; Torres-Álvarez, B. Near-visible light and UV photoprotection in the treatment of melasma: A double-blind randomized trial. Photodermatol. Photoimmunol. Photomed. 2014, 30, 35–42. [Google Scholar] [CrossRef]
  196. Kaye, E.T.; Levin, J.A.; Blank, I.H.; Arndt, K.A.; Anderson, R.R. Efficiency of opaque photoprotective agents in the visible light range. Arch. Dermatol. 1991, 127, 351–355. [Google Scholar] [CrossRef]
  197. Rodrigues, M.; Pandya, A.G. Melasma: Clinical diagnosis and management options. Australas. J. Dermatol. 2015, 56, 151–163. [Google Scholar] [CrossRef]
  198. Victor, F.C.; Gelber, J.; Rao, B. Melasma: A review. J. Cutan. Med. Surg. Inc. Med. Surg. Dermatol. 2004, 8, 97–102. [Google Scholar] [CrossRef]
  199. Osterwalder, U.; Sohn, M.; Herzog, B. Global state of sunscreens. Photodermatol. Photoimmunol. Photomed. 2014, 30, 62–80. [Google Scholar] [CrossRef] [PubMed]
  200. Khanna, V.J.; Shieh, S.; Benjamin, J.; Somach, S.; Zaim, M.T.; Dorner, W., Jr.; Shill, M.; Wood, G.S. Necrolytic acral erythema associated with hepatitis C: Effective treatment with interferon alfa and zinc. Arch. Dermatol. 2000, 136, 755–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Abdallah, M.A.; Hull, C.; Horn, T.D. Necrolytic Acral Erythema: A Patient From the United States Successfully Treated With Oral Zinc. Arch. Dermatol. 2005, 141, 85–87. [Google Scholar] [CrossRef] [PubMed]
  202. Najarian, D.J.; Lefkowitz, I.; Balfour, E.; Pappert, A.S.; Rao, B.K. Zinc deficiency associated with necrolytic acral erythema. J. Am. Acad. Dermatol. 2006, 55 (Suppl. 5), S108–S110. [Google Scholar] [CrossRef] [PubMed]
  203. Sinclair, S.A.; Reynolds, N.J. Necrolytic migratory erythema and zinc deficiency. Br. J. Dermatol. 1997, 136, 783–785. [Google Scholar] [CrossRef]
  204. Sharquie, K.E.; Najim, R.A.; Al-Hayani, R.K.; Al-Nuaimy, A.A.; Maroof, D.M. The therapeutic and prophylactic role of oral zinc sulfate in management of recurrent aphthous stomatitis (ras) in comparison with dapsone. Saudi Med. J. 2008, 29, 734–738. [Google Scholar]
  205. Altenburg, A.; El-Haj, N.; Micheli, C.; Puttkammer, M.; Abdel-Naser, M.B.; Zouboulis, C.C. The treatment of chronic recurrent oral aphthous ulcers. Dtsch. Arztebl. Int. 2014, 111, 665–673. [Google Scholar] [CrossRef] [Green Version]
  206. Belenguer-Guallar, I.; Jiménez-Soriano, Y.; Claramunt-Lozano, A. Treatment of recurrent aphthous stomatitis. A literature review. J. Clin. Exp. Dent. 2014, 6, e168. [Google Scholar] [CrossRef]
  207. Edgar, N.R.; Saleh, D.; Miller, R.A. Recurrent aphthous stomatitis: A review. J. Clin. Aesthetic Dermatol. 2017, 10, 26. [Google Scholar]
  208. Skaare, A.B.; Herlofson, B.B.; Barkvoll, P. Mouthrinses containing triclosan reduce the incidence of recurrent aphthous ulcers (RAU). J. Clin. Periodontol. 1996, 23, 778–781. [Google Scholar] [CrossRef] [PubMed]
  209. Mehdipour, M.; Taghavi Zenouz, A.; Bahramian, A.; Yazdani, J.; Pouralibaba, F.; Sadr, K. Comparison of the Effect of Mouthwashes with and without Zinc and Fluocinolone on the Healing Process of Erosive Oral Lichen Planus. J. Dent. Res. Dent. Clin. Dent. Prospect. 2010, 4, 25–28. [Google Scholar]
  210. Chaitanya, N.C.; Chintada, S.; Kandi, P.; Kanikella, S.; Kammari, A.; Waghamare, R.S. Zinc Therapy in Treatment of Symptomatic Oral Lichen Planus. Indian Dermatol. Online J. 2019, 10, 174–177. [Google Scholar] [CrossRef] [PubMed]
  211. Faergemann, J.; Fredriksson, T. An open trial of the effect of a zinc pyrithione shampoo in tinea versicolor. Cutis 1980, 25, 667–669. [Google Scholar] [PubMed]
  212. Gupta, A.K.; Foley, K.A. Antifungal Treatment for Pityriasis Versicolor. J. Fungi 2015, 1, 13–29. [Google Scholar] [CrossRef]
  213. Hald, M.; Arendrup, M.C.; Svejgaard, E.L.; Lindskov, R.; Foged, E.K.; Saunte, D.M. Evidence-based Danish guidelines for the treatment of Malassezia-related skin diseases. Acta Derm.-Venereol. 2015, 95, 12–19. [Google Scholar] [CrossRef] [Green Version]
  214. Sharquie, K.E.; Al-Dori, W.S.; Sharquie, I.K.; Al-Nuaimy, A.A. Treatment of Pityriasis Versicolour with Topical 15% Zinc Sulfate Solution. raqi J. Comm. Med. 2008, 21, 61–63. [Google Scholar]
  215. Sadeghian, G.; Ziaei, H.; Nilforoushzadeh, M. Treatment of localized psoriasis with a topical formulation of zinc pyrithione. Acta Derm. APA 2011, 20, 187–190. [Google Scholar]
  216. Clemmensen, O.J.; Siggaard-Andersen, J.; Worm, A.M.; Stahl, D.; Frost, F.; Bloch, I. Psoriatic arthritis treated with oral zinc sulphate. Br. J. Dermatol. 1980, 103, 411–415. [Google Scholar] [CrossRef]
  217. Strömberg, H.-E.; Ågren, M.S. Topical zinc oxide treatment improves arterial and venous leg ulcers. Br. J. Dermatol. 1984, 111, 461–468. [Google Scholar] [CrossRef]
  218. Ågren, M.S.; Strömberg, H.-E. Topical Treatment of Pressure Ulcers: A Randomized Comparative Trial of Varidase® and Zinc Oxide. Scand. J. Plast. Reconstr. Surg. 1985, 19, 97–100. [Google Scholar] [CrossRef] [PubMed]
  219. Apelqvist, J.; Larsson, J.; Lstenström, A. Topical treatment of necrotic foot ulcers in diabetic patients: A comparative trial of DuoDerm and MeZinc. Br. J. Dermatol. 1990, 123, 787–792. [Google Scholar] [CrossRef] [PubMed]
  220. Cornwall, M.W. Zinc iontophoresis to treat ischemic skin ulcers. Phys. Ther. 1981, 61, 359–360. [Google Scholar] [CrossRef] [PubMed]
  221. Yaghoobi, R.; Omidian, M.; Bagherani, N. Comparison of therapeutic efficacy of topical corticosteroid and oral zinc sulfate-topical corticosteroid combination in the treatment of vitiligo patients: A clinical trial. BMC Dermatol. 2011, 11, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Sharquie, K.E.; Khorsheed, A.A.; Al-Nuaimy, A.A. Topical zinc sulphate solution for treatment of viral warts. Saudi Med. J. 2007, 28, 1418–1421. [Google Scholar] [PubMed]
  223. Mun, J.-H.; Kim, S.-H.; Jung, D.-S.; Ko, H.-C.; Kim, B.-S.; Kwon, K.-S.; Kim, M.-B. Oral zinc sulfate treatment for viral warts: An open-label study. J. Dermatol. 2011, 38, 541–545. [Google Scholar] [CrossRef]
  224. Yaghoobi, R.; Sadighha, A.; Baktash, D. Evaluation of oral zinc sulfate effect on recalcitrant multiple viral warts: A randomized placebo-controlled clinical trial. J. Am. Acad. Dermatol. 2009, 60, 706–708. [Google Scholar] [CrossRef]
  225. Khattar, J.A.; Musharrafieh, U.M.; Tamim, H.; Hamadeh, G.N. Topical zinc oxide vs. salicylic acid–lactic acid combination in the treatment of warts. Int. J. Dermatol. 2007, 46, 427–430. [Google Scholar] [CrossRef]
  226. Friedland, B.A.; Hoesley, C.J.; Plagianos, M.; Hoskin, E.; Zhang, S.; Teleshova, N.; Alami, M.; Novak, L.; Kleinbeck, K.R.; Katzen, L.L.; et al. First-in-Human Trial of MIV-150 and Zinc Acetate Coformulated in a Carrageenan Gel: Safety, Pharmacokinetics, Acceptability, Adherence, and Pharmacodynamics. J. Acquir. Immune Defic. Syndr. 2016, 73, 489–496. [Google Scholar] [CrossRef]
  227. De Laat, J.T.; De Gruijl, F. The role of UVA in the aetiology of non-melanoma skin cancer. Cancer Surv. 1996, 26, 173–191. [Google Scholar]
  228. Stary, A.; Robert, C.; Sarasin, A. Deleterious effects of ultraviolet A radiation in human cells. Mutat. Res./DNA Repair 1997, 383, 1–8. [Google Scholar] [CrossRef] [PubMed]
  229. Danpure, H.J.; Tyrrell, R.M. Oxygen-dependence of near UV (365 NM) lethality and the interaction of near UV and X-rays in two mammalian cell lines. Photochem. Photobiol. 1976, 23, 171–177. [Google Scholar] [CrossRef] [PubMed]
  230. Mohammed, Y.H.; Barkauskas, D.S.; Holmes, A.; Grice, J.; Roberts, M.S. Noninvasive in vivo human multiphoton microscopy: A key method in proving nanoparticulate zinc oxide sunscreen safety. J. Biomed. Opt. 2020, 25, 014509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Leccia, M.-T.; Richard, M.-J.; Favier, A.; B’Eani, J.-C. Zinc protects against ultraviolet A1-induced DNA damage and apoptosis in cultured human fibroblasts. Biol. Trace Elem. Res. 1999, 69, 177–190. [Google Scholar] [CrossRef] [PubMed]
  232. Lewicka, Z.A.; Yu, W.W.; Oliva, B.L.; Contreras, E.Q.; Colvin, V.L. Photochemical behavior of nanoscale TiO2 and ZnO sunscreen ingredients. J. Photochem. Photobiol. A Chem. 2013, 263, 24–33. [Google Scholar] [CrossRef]
  233. Mitchnick, M.A.; Fairhurst, D.; Pinnell, S.R. Microfine zinc oxide (Z-Cote) as a photostable UVA/UVB sunblock agent. J. Am. Acad. Dermatol. 1999, 40, 85–90. [Google Scholar] [CrossRef]
  234. Pinnell, S.R.; Fairhurst, D.; Gillies, R.; Mitchnick, M.A.; Kollias, N. Microfine Zinc Oxide is a Superior Sunscreen Ingredient to Microfine Titanium Dioxide. Dermatol. Surg. 2000, 26, 309–314. [Google Scholar] [CrossRef]
  235. Adler, B.L.; DeLeo, V.A. Sunscreen Safety: A Review of Recent Studies on Humans and the Environment. Curr. Dermatol. Rep. 2020, 9, 1–9. [Google Scholar] [CrossRef]
  236. Mancuso, J.B.; Maruthi, R.; Wang, S.Q.; Lim, H.W. Sunscreens: An Update. Am. J. Clin. Dermatol. 2017, 18, 643–650. [Google Scholar] [CrossRef]
  237. Schneider, S.L.; Lim, H.W. A review of inorganic UV filters zinc oxide and titanium dioxide. Photodermatol. Photoimmunol. Photomed. 2019, 35, 442–446. [Google Scholar] [CrossRef]
  238. Mohammed, Y.H.; Holmes, A.; Haridass, I.N.; Sanchez, W.Y.; Studier, H.; Grice, J.E.; Benson, H.A.E.; Roberts, M.S. Support for the Safe Use of Zinc Oxide Nanoparticle Sunscreens: Lack of Skin Penetration or Cellular Toxicity after Repeated Application in Volunteers. J. Investig. Dermatol. 2019, 139, 308–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Monteiro-Riviere, N.A.; Wiench, K.; Landsiedel, R.; Schulte, S.; Inman, A.O.; Riviere, J.E. Safety evaluation of sunscreen formulations containing titanium dioxide and zinc oxide nanoparticles in UVB sunburned skin: An in vitro and in vivo study. Toxicol. Sci. 2011, 123, 264–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Cross, S.E.; Innes, B.; Roberts, M.S.; Tsuzuki, T.; Robertson, T.A.; McCormick, P. Human skin penetration of sunscreen nanoparticles: In-vitro assessment of a novel micronized zinc oxide formulation. Ski. Pharmacol. Physiol. 2007, 20, 148–154. [Google Scholar] [CrossRef] [PubMed]
  241. Mann, J.; Truswell, A.S. Essentials of Human Nutrition; Oxford University Press: Oxford, UK, 2017. [Google Scholar]
  242. Plum, L.M.; Rink, L.; Haase, H. The essential toxin: Impact of zinc on human health. Int. J. Environ. Res. Public Health 2010, 7, 1342–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. European Commission, Scientific Committee on Consumer Safety. Opinion on Zinc Oxide (Nano Form): COLIPA n° S76; European Commission: Brussels, Belgium, 2013. [Google Scholar]
  244. Ginzburg, A.L.; Blackburn, R.S.; Santillan, C.; Truong, L.; Tanguay, R.L.; Hutchison, J.E. Zinc oxide-induced changes to sunscreen ingredient efficacy and toxicity under UV irradiation. Photochem. Photobiol. Sci. 2021, 20, 1273–1285. [Google Scholar] [CrossRef] [PubMed]
  245. Corinaldesi, C.; Marcellini, F.; Nepote, E.; Damiani, E.; Danovaro, R. Impact of inorganic UV filters contained in sunscreen products on tropical stony corals (Acropora spp.). Sci. Total Environ. 2018, 637–638, 1279–1285. [Google Scholar] [CrossRef]
Figure 1. Anatomy of the integumentary system showing different structures within the epidermis, dermis, and hypodermis.
Figure 1. Anatomy of the integumentary system showing different structures within the epidermis, dermis, and hypodermis.
Ijms 23 16165 g001
Figure 2. Zinc transporters SLC39A (ZIP) and SLC30A (ZnT) with Zn2+ ion transportation direction. ZIP is predicted to have 8 transmembrane domains and ZnT is predicted to have 6.
Figure 2. Zinc transporters SLC39A (ZIP) and SLC30A (ZnT) with Zn2+ ion transportation direction. ZIP is predicted to have 8 transmembrane domains and ZnT is predicted to have 6.
Ijms 23 16165 g002
Table 1. Therapeutic applications of zinc in the treatment of various skin disorders.
Table 1. Therapeutic applications of zinc in the treatment of various skin disorders.
DisorderEtiologyTreatmentReferences
Acne conglobataPropionibacterium acnesSuccessfully treated with a high dose of oral zinc sulphate.[152]
Acne vulgarisPropionibacterium acnesClindamycin or erythromycin in combination with zinc acetate or octoate was found to boost therapy efficacy.[145,146,150,153,154,155,156,157,158,159,160,161,162]
Papular and pustular acne can be cured with oral zinc sulphate.
Oral zinc gluconate is effective in the management of inflammatory acne.
Antioxidants combined with methionine-bound zinc complex was successful in treating mild to moderate conditions.
The alternative route of treatment can be zinc alone or in combination with nicotinamide.
Actinic keratosisUV exposureTopical 25% zinc sulphate resulted in the disappearance of the lesions.[163]
Alopecia areataautoimmune disorderOral zinc supplementation showed a noticeable clinical response.[164,165]
Athlete’s footTrichophyton rubrum20% zinc-undecylenate-containing powder was found to be effective in reducing erythema, scaling, and itching.[166,167]
Androgenetic alopeciaandrogens, genetic predispositionSignificant hair growth was observed with topical zinc pyrithione 1% solution.[168,169]
Behcet’s diseaseautoimmune disorderBehcet’s disease was treated with oral zinc sulphate.[170,171]
BromhidrosisCorynebacterium sp.Topical zinc salt such as sulphate and zinc oxide were found to be successful in the management of the condition.[172,173]
BromodosisSweat buildup leading to bacterial or fungal growthA topical 15% zinc sulphate solution was found to eliminate foot odor.[174]
Cutaneous leishmaniasisLeishmaniaIntralesional 2% zinc sulphate with meglumine and oral zinc sulphate was found to be effective in the management of cutaneous leishmaniasis.[175,176,177]
Dissecting cellulitis of the scalpUnknownComplete cure with oral zinc sulphate.[152,178,179]
EczemaImmune system overactivityTextiles treated with zinc oxide can be useful in the management of atopic dermatitis.[180,181,182]
For diaper dermatitis, zinc oxide paste was found to be effective in soothing and preventing skin rash.
For hand eczema, a cream containing zinc sulphate (2.5%) combined with clobetasol (0.05%) has improved the condition.
Erosive pustular dermatosis of the scalpUnknownTreated with oral zinc sulphate.[183]
Herpes genitalisHerpes simplex virus type 2Zinc acetate gel was effective in the prevention of sexual transmission of HSV-2 and HIV.[184,185]
Higher concentrations of zinc sulphate were found more effective in the treatment, and prevention of relapse.
Herpes simplexHerpes simplex virus type 1Polyethylene glycol-coated zinc oxide nanoparticles demonstrated antiviral potency against HSV-1[186,187]
Zinc gluconate and zinc lactate was found to effectively inactivate HSV-1 clinical isolates.
Hidradenitis
suppurativa
UnknownThe disorder can be managed with oral zinc gluconate alone or in combination with topical triclosan.[188,189]
Jock itchTrichophyton rubrum
Trichophyton mentagrophytes
A cream formulated with 20% zinc undecylenate has effectively cleared the skin.[166,190]
LeprosyMycobacterium lepraeCombining oral zinc with dapsone was found to enhance the therapy’s effectiveness through bacterial clearance and rapid conversion of lepromin.[191,192,193]
Topical application of phenytoin sodium zinc oxide paste showed a significant clearance of the bacterial load of trophic ulcers.
MelasmaUV exposureTopical 10% zinc sulphate resulted in a significant decrease in MASI* scores.[194,195,196,197,198,199]
Zinc oxide in sunscreen formulations is used in the management of melasma owing to its photoprotection properties.
Necrolytic acral erythemaAssociated with hepatitis CThe condition was treated with oral zinc supplementation.[200,201,202]
Necrolytic migratory erythemaAssociated with pancreatic glucagonomaOral zinc sulfate has been shown to improve the condition.[203]
Oral aphthous stomatitisUnknownOral zinc sulphate lowered the risk of relapse in recurrent aphthae and provided both curative and preventative effects.[204,205,206,207,208]
Zinc sulphate-containing mouth rinse decreased the frequency of recurring ulcers.
Oral lichen planusUnknown0.2% zinc mouthwash in combination with fluocinolone helped diminish irritability, pain, and lesion surface area.[209,210]
Administration of oral zinc acetate showed favorable clinical improvement.
Pityriasis versicolorMalasseziaZinc pyrithione 1% in shampoo formulations was found effective in the treatment of pityriasis versicolor.[211,212,213,214]
Topical 15% zinc sulphate was effective in the treatment of pityriasis versicolor.
PsoriasisUnknownTopical 0.25% zinc pyrithione was found effective for localized plaque psoriasis.[215]
Psoriatic arthritisUnknownPsoriatic arthritis can be effectively treated with oral zinc sulphate.[148,216]
Seborrheic dermatitisMalasseziaZinc pyrithione 1% in a shampoo formulation is a therapeutic choice for reducing inflammations and scaling.[215]
UlcersPoor blood flowTopical zinc oxide formulations have been used in the treatment of arterial and venous leg ulcers, pressure ulcers, and diabetic foot ulcers.[6,217,218,219,220]
Zinc iontophoresis was demonstrated to be beneficial in the treatment of ischemic skin ulcers.
VitiligoMelanocyte decrement in relation to genetic and non-genetic factorsOral zinc sulphate in combination with topical corticosteroids showed a higher response rate than Topical corticosteroids alone in the treatment of vitiligo.[221]
WartsHuman papillomavirusTopical 10% zinc sulfate was found effective for the treatment of plane warts[222,223,224,225,226]
Oral zinc sulfate can be used in the treatment of different types of warts.
Topical 20% zinc oxide is considered an effective and safe therapeutic method.
Zinc acetate coformulated in a carrageenan gel demonstrated anti-HIV and anti-human papillomavirus activity.
MASI*: melasma area and severity index.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Al-Khafaji, Z.; Brito, S.; Bin, B.-H. Zinc and Zinc Transporters in Dermatology. Int. J. Mol. Sci. 2022, 23, 16165. https://doi.org/10.3390/ijms232416165

AMA Style

Al-Khafaji Z, Brito S, Bin B-H. Zinc and Zinc Transporters in Dermatology. International Journal of Molecular Sciences. 2022; 23(24):16165. https://doi.org/10.3390/ijms232416165

Chicago/Turabian Style

Al-Khafaji, Zubaidah, Sofia Brito, and Bum-Ho Bin. 2022. "Zinc and Zinc Transporters in Dermatology" International Journal of Molecular Sciences 23, no. 24: 16165. https://doi.org/10.3390/ijms232416165

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop