Next Article in Journal
Mechanotransduction-Driven Modulation of L-Type Calcium Channels: Roles of Nitric Oxide, S-Nitrosylation, and cGMP in Rat Ventricular Cardiomyocytes
Previous Article in Journal
Data Mining and Biochemical Profiling Reveal Novel Biomarker Candidates in Alzheimer’s Disease
Previous Article in Special Issue
Review of Applications of Zeolites in Dermatology: Molecular Perspectives and Translational Potentials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Perspectives on Nutraceutical Insulin Sensitizing Agents in the Treatment of Psoriasis and Other Dermatological Diseases

1
Outpatient Specialist in Dermatology, Azienda Sanitaria Provinciale, 87100 Cosenza, Italy
2
Unit of Dermatology, Department of Medicine, Dimed, University of Padova, 35128 Padova, Italy
3
Regional Center of Pediatric Dermatology, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy
4
Gynecology and Obstetrics, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
5
Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy
6
CAOM—Centro Studi Applicati Erbe Officinali e Frutti Minori, 28100 Novara, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(15), 7538; https://doi.org/10.3390/ijms26157538
Submission received: 14 June 2025 / Revised: 25 July 2025 / Accepted: 29 July 2025 / Published: 4 August 2025

Abstract

Insulin resistance (IR) plays a pivotal role in the pathogenesis of several dermatological diseases, including psoriasis, acne, acanthosis nigricans, and hidradenitis suppurativa (HS). These conditions are characterized by chronic inflammation, oxidative stress, and metabolic dysfunction, which are exacerbated by IR. This narrative review examines the emerging role of nutraceutical insulin-sensitizing agents (ISAs), including myo-inositol, alpha-lipoic acid, vitamin D, vitamin C, and folic acid, in managing IR-related dermatological disorders. A comprehensive literature search was conducted across Cochrane Library and MEDLINE (1965–May 2025), focusing on clinical trials involving nutraceutical ISAs in dermatological conditions associated with IR. Only human studies published in English were included. Evidence from randomized controlled trials (RCTs) and observational studies suggests that ISAs improve glycemic control, reduce oxidative stress, and modulate inflammatory pathways in IR-related dermatoses. Notably, myo-inositol combined with magnesium and folic acid has demonstrated significant reductions in acne severity, hirsutism, and quality-of-life impairments in women with polycystic ovary syndrome. Similar benefits have been observed in psoriasis and HS, though data remain limited. Nutraceutical ISAs offer a promising adjunctive approach for the management of IR-associated dermatological diseases, potentially addressing both metabolic dysfunction and skin inflammation. However, robust RCTs with long-term follow-up are needed to confirm these preliminary findings and to establish optimal treatment regimens.

1. Introduction

Identified in 1910 by Sir Edward Albert Sharpey-Shafer, insulin was initially recognized as the crucial substance missing in individuals with type 1 diabetes (T1D) [1]. It has since become one of the most groundbreaking discoveries in medical science during the 20th century, revolutionizing treatment strategies and significantly improving the life expectancy of diabetic individuals. Beyond its well-established function in glucose regulation, insulin is now acknowledged as a growth factor and a vital biological modulator that participates in various physiological processes across all tissues [2]. As scientific understanding of insulin’s metabolic role continues to grow, so does awareness of the consequences that arise from disturbances in its signaling pathways, shedding light on the pathogenic mechanisms underlying numerous diseases. Insulin, which is secreted by pancreatic beta cells, plays a central role in glucose homeostasis and also influences cellular processes such as proliferation, differentiation, and apoptosis. Insulin resistance (IR) occurs when cells become less responsive to insulin, resulting in hyperinsulinemia, heightened androgen synthesis, and metabolic irregularities. Insulin resistance (IR) is known to trigger pro-inflammatory signaling cascades, including the activation of the mitogen-activated protein kinase (MAPK) pathway and phosphoinositide 3-kinase (PI3-K) cascades, which in turn disrupt skin homeostasis [3].
Several dermatological conditions, including acanthosis nigricans, acne, psoriasis, hidradenitis suppurativa, androgenetic alopecia, and hirsutism, have been linked to IR [4]. Under normal circumstances, insulin helps maintain a balance between keratinocyte proliferation and differentiation, both of which are crucial for the proper formation of the epidermal structure. However, in chronic inflammatory conditions like acne and psoriasis, elevated levels of pro-inflammatory cytokines activate the p38MAPK pathway, exacerbating IR by inducing serine phosphorylation of insulin receptor substrates (IRSs). This modification impairs keratinocyte differentiation while simultaneously promoting basal proliferation of these cells (Figure 1) [4].
The growing body of evidence surrounding the relationship between IR and skin disorders has significant therapeutic implications. There is increasing support for the use of insulin-sensitizing agents (ISAs) in managing a range of medical conditions now recognized as being linked to IR.
This review aims to explore the potential role of nutraceutical ISAs in the management of dermatological diseases associated with IR, including psoriasis, acne, acanthosis nigricans, and hidradenitis suppurativa. Although the connection between IR and metabolic disorders has been extensively studied, its relevance in dermatology remains underappreciated. This review adopts a translational and mechanistic framework that integrates the pathophysiological underpinnings of IR with emerging clinical evidence on the application of ISAs in dermatology. Through a dual focus on molecular mechanisms and interventional studies, the authors offer a comprehensive and up-to-date synthesis of how specific nutraceuticals, such as myo-inositol, alpha-lipoic acid, vitamin D, vitamin C, and folic acid, may modulate inflammatory and metabolic signaling cascades implicated in cutaneous disorders.

2. Overview of Insulin Signaling and Resistance Mechanisms

By regulating glucose uptake and gluconeogenesis, insulin plays a crucial role in glucose homeostasis through a complex and systemic signaling. To enhance blood glucose supply to target organs, insulin promotes the release of nitric oxide (NO) and endothelin in the endothelium, leading to vasodilation and vasoconstriction, respectively [5]. Furthermore, insulin reduces the expression of gluconeogenic enzymes in hepatocytes and boosts glycogen synthesis and enhances glucose uptake through the glucose transporter type 4 (GLUT-4) receptor in adipocytes and skeletal myocytes [6]. Moreover, insulin suppresses orexigenic neuropeptides, i.e., neuropeptide Y and agouti-related peptide in the hypothalamus, while it increases anorexigenic NP such as cocaine- and amphetamine-regulated transcript and proopiomelanocorticotropin in the arcuate nucleus [7]; this increases the activity of the α-Melanocyte-Stimulating Hormone in the paraventricular nucleus to decrease food intake [8]. By binding to its cell membrane receptor, insulin triggers autophosphorylation of intracellular tyrosine residues, resulting in the recruitment and phosphorylation of IRSs, i.e., IRS1 and IRS2 [6]. Two main intracellular pathways are described and simplified as follows:
The phosphoinositide 3-kinase (PI3K) pathway, also known as the protein kinase B (PKB or Akt) pathway, facilitates GLUT-4 translocation to the membrane, glucose transport into the cell, and glycogen synthesis. This brings about the activation of PI3K by converting phosphatidyl inositol 4,5-biphosphate (PIP2) into phosphatidyl inositol 3,4,5-triphosphate (PIP3), the recruitment of Akt to the plasma membrane, and its phosphorylation by 3-phosphoinositide-dependent kinase-1 (PDK-1) and the mammalian target of rapamycin complex 2 (mTORC-2) [9]. This pathway results in the translocation of GLUT-4 in adipocytes and skeletal myocytes and the activation of glycogen synthase (GS) in skeletal myocytes, adipocytes, hepatocytes, and endothelial NO synthase (NOS) in endothelial cells, along with the phosphorylation of transcription factor forkhead box protein O1 (FoxO1) to suppress hepatic gluconeogenesis [9].
The MAPK pathway, also known as the extracellular signal regulated kinase (ERK) pathway, controls transcription factors involved in cell growth, differentiation, and proliferation, while entailing the activation of ERK and the synthesis of endothelin-1 in endothelial cells [10].
These molecular mechanisms (Figure 2) rely on a very delicate biochemical balance that is affected by homeostasis impairment. Inhibitory kinases which alter glucose assimilation and glycogen synthesis while stimulating gluconeogenesis by phosphorylation of IRS-1/2 are activated in the presence of inflammatory cytokines, oxidative stress (OS), and free fatty acids (FFAs) [11]. Moreover, several metabolites such as diacylglycerol and ceramides affect the modulation of insulin signaling [12]. In IR, the signaling pathways are impaired and cells do not respond appropriately to normal levels of insulin. A number of intrinsic and extrinsic factors contribute to the development of IR, including obesity, stress, and aging [13]. In the presence of IR, mitochondria are reduced in number and their morphology is disrupted with a decrease in ATP synthesis [14]. This metabolic vicious circle leads to an increase in reactive oxygen species (ROS), while greater serine phosphorylation of IRS-1/2 inhibits the insulin signaling pathway downstream, ultimately leading to an increased glucose production along with its reduced uptake, vasodilation, and insulin secretion [15]. Several additional cellular mechanisms have been recognized as potential contributors to the development of IR both in vivo and in vitro, highlighting that this condition is characterized by an overall inflamed and dysfunctional cell environment [16]. In this context, the role of biogenic amines, such as histamine, tyramine, cadaverine, and putrescine, is gaining increasing attention. These amines, produced endogenously or by gut microbiota, have been shown to modulate insulin signaling and contribute to systemic inflammation. In particular, histamine can promote pro-inflammatory cytokine release and oxidative stress, both of which impair insulin receptor function. Moreover, certain biogenic amines may influence sebaceous gland activity and hormonal balance, establishing a potential mechanistic link between insulin resistance, androgen excess, and acne vulgaris [17].
Moreover, insulin resistance (IR) is associated with decreased levels of sex hormone-binding globulin (SHBG), elevated concentrations of luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and a subsequent increase in ovarian androgen synthesis, which may contribute to the development of hyperandrogenism [18,19].
IR is often described alongside the broader metabolic syndrome (MetS) that includes cardiovascular risk factors, i.e., hypertension, central obesity, impaired glucose tolerance, and dyslipidemia, which may cause non-alcoholic fatty liver disease (NAFLD) which can later progress into non-alcoholic steatohepatitis (NASH) [20]. The prevalence of IR is lowest among European adults (15.5%), while it has been reported to be as high as 23.3% in Thailand, 39.1% in Texas (US), and 46.5% in Venezuela [21,22]. Age plays a crucial role in the development of IR, since increased OS and mitochondrial dysfunction typically occur as a result of the aging process [21]. Gender must also be considered in the pathogenesis of IR as adult men normally have more visceral and hepatic adipose tissue than female subjects; this increases the presence of FFAs and pro-inflammatory cytokines that, along with the lack of the protective effect of estrogen and lower adiponectin levels, makes males more prone to IR than pre-menopausal women [21].
Besides its role in dermatoses, which is the topic of the present publication, IR plays a pivotal role in several conditions such as overactive bladder (OAB), fibromyalgia (FM), polycystic ovary syndrome (PCOS), and rheumatoid arthritis (RA), which are characterized by chronic inflammation mechanisms and not rarely associated as comorbidities by pathogenetic loops which are still not completely understood [23].

3. IR in Dermatological Conditions

As the largest of our organs, the skin may show signs of a serious and underlying internal disease, e.g., diabetes, lupus erythematosus, inflammatory bowel diseases, systemic sclerosis, and some types of cancer [24]. Among its many actions, insulin also affects the homeostasis and physiology of the skin as it regulates the balance between proliferation and differentiation of keratinocytes, which is a prerequisite for the formation of a healthy epidermal structure [25]. However, the highly inflamed environment which features IR leads to the activation of p38MAPK and serine phosphorylation of IRS, with a subsequent blockade of differentiation and increased proliferation of basal keratinocytes [26]. Therefore, IR is associated with pathogenesis and the severity of a few dermatological conditions which are not rarely linked to some conditions associated with overall dysmetabolism.

3.1. Psoriasis

Psoriasis is a systemic chronic immunomediated disorder featuring papulo-squamous plaques which consist of salmon to pink lesions covered by silvery scales, which are usually distributed symmetrically on the extensor aspects of the elbows and knees, scalp, and/or lumbosacral region [27]. However, psoriasis is an extremely complex condition which can display other skin symptoms and concomitant debilitating arthritis [28]. While obesity and overweight are considered risk and exacerbating factors for psoriasis itself, psoriatic patients are at high risk of developing metabolic diseases including type 2 diabetes (T2D), MetS, and cardiovascular conditions [29,30,31]. In particular, subjects with psoriatic arthritis (PsA) show a severe endothelial dysfunction which is directly correlated to their homeostasis model assessment (HOMA)-IR outcomes which put them at high risk of developing cardiovascular atherosclerotic comorbidities [32]. Psoriasis and metabolic diseases share an inflammatory pathogenesis, and adipose tissue (AT) is now recognized to play a role in this connection [33]. Adipocytokines such as leptin and adiponectin, which regulate and affect insulin sensitivity and signaling, are deregulated in both psoriasis and obesity, and plasma levels of adiponectin, which has anti-inflammatory action, are decreased in obesity, psoriasis, IR, and T2D [34]. Moreover, these adipokines also regulate several immune functions such as cytokine production and T-cell differentiation, emphasizing the intricate relationship between immune and metabolic dysfunctions and further supporting the link between psoriasis and IR [33]. Other adipocytokines such as omentin, visfatin, and resistin, which play an important role in insulin sensitivity, were found to be altered in psoriatics [35]. Tumor necrosis factor-α (TNF-α), a key cytokine in the pathogenesis of psoriasis, impairs insulin signaling by inhibiting the tyrosine kinase activity of the insulin receptor and reducing adiponectin secretion [36]. The association between psoriasis and IR is further supported by clinical evidence: several studies have reported significantly reduced insulin sensitivity in patients with psoriasis compared to healthy controls [37]; moreover, both serum insulin levels and IR indices positively correlate with disease severity [38]. In glucose-tolerant individuals, moderate to severe psoriasis has been linked to a marked reduction in insulin sensitivity when compared with non-psoriatic counterparts [39]. Further data have reinforced the connection between IR and psoriasis by showing a significantly higher prevalence of conditions which are normally related to IR, hyperinsulinemia, and dyslipidemia such as PCOS in psoriatic women than in healthy controls [40,41].

3.2. Acne

Acne is an inflammatory condition of the folliculo-pilosebaceous unit, characterized by comedones, papules, pustules, or nodules which are generally located on the face, shoulders, back, and chest [42]. Acne features a complex etiopathology which includes hyperseborrhea, hyperkeratosis, and inflammation, and it seems to be strongly associated with IR [41]. Many transcription factors such as FoxO1, 1,25-dihydroxyvitamin D and calcium are connected with sebum production, while hyperandrogenemia, hyperinsulinemia, and high levels of insulin growth factor-1 (IGF-1) play a role in acne development [43,44]. In particular, hyperinsulinemia increases keratinocytes proliferation and dysfunction by triggering IGF-1 receptors, which leads to sebocyte hyperproliferation [45]. Moreover, IGF-1 enhances androgen receptor signal transduction, elevating androgen levels and causing hyperseborrhea [46]. Increased serum insulin/IGF-1 significantly correlates with a higher dietary glycaemic load, which is a common finding in acne patients, activates mTORC1, causing IR via ribosomal protein S6 kinase b1 (S6K1), and inhibits FoxO1 which represses androgen signaling, resulting in sebaceous gland dysfunction [47,48]. Moreover, a positive correlation between a higher body mass index (BMI) and acne severity has been observed [49]. On the contrary, a lower dietary glycaemic load decreases inflammation, pro-inflammatory chemokines, and the size of sebaceous glands [24]. High glycaemia induces hyperinsulinemia and increases androgen production, while lowering the serum levels of SHBG, intensifying androgen activity, and facilitating acne development [50,51]. In patients with acne, increased mTORC1 activity has been detected, which is strongly associated with IR, obesity, T2D, and cancers such as melanoma [46]; moreover, a significant correlation between decreased expression of insulin, IGF-1, and mTORC1 and a reduced prevalence rate of acne has been observed [41]. In addition, a few studies have pointed out a direct correlation between HOMA-IR values and acne development [52].
Recent evidence also suggests that IR exacerbates oxidative stress, which plays a key role in acne pathogenesis [53]. In addition, insulin-resistant states increase ROS production in the skin, impairing keratinocyte differentiation, promoting lipid peroxidation, and activating pro-inflammatory pathways such as NF-κB and MAPK [54]. This oxidative microenvironment contributes to cutaneous inflammation and may also favor dysbiosis, increasing Cutibacterium acnes proliferation and aggravating acne severity [55]. These findings highlight the pathogenic synergy between metabolic dysfunction, redox imbalance, and inflammatory signaling in acne.

3.3. Acanthosis Nigricans

Acanthosis nigricans (AN) is characterized by velvety, hyperpigmented skin plaques typically located in areas such as the neck, axilla, and knuckles, as well as the groin, umbilical, and perianal regions [56]. Obesity-associated AN is the most common disease subtype and it is linked to IR and hyperinsulinemia [57]. Indeed, hyperinsulinemia stimulates IGF-receptors with subsequent keratinocyte proliferation [58]. The activity of IGF-1 is regulated by IGF binding proteins (IGFBPs) which increase IGF-1 half life, and IGFBP-1 and IGFBP-2 are both decreased in obese subjects with hyperinsulinemia, increasing bioactive free IGF-1 which promotes cell growth and differentiation, thereby facilitating the development of hyperkeratosis and papillomatosis observed in AN [59]. The current prevalence of AN ranges from 4.5 to 74%, and an alarming escalation of cases has been noted in younger populations affected by obesity and MetS [60,61,62]. In addition, AN has been reported as a clinical manifestation in subjects affected by Down syndrome, who are prone to obesity, MetS, and T2D [63]. In patients with AN, IR-associated skin lesions are usually located in the neck, axilla, and knuckles, even though unusual locations such as the face have been described. AN has been associated with the skin symptoms of T1D, T2D, and also PCOS [64]. Moreover, AN has been suggested as a useful clinical marker of IR particularly applicable for screening on a larger scale, such as in obese children and teenagers, populations highly susceptible to T2D and MetS, and subjects diagnosed with prediabetes status and dyslipidemia [65].

3.4. Hidradenitis Suppurativa

Hidradenitis suppurativa (HS) is a chronic skin disorder characterized by recurrent abscesses, draining, and scarring which involves the terminal follicular acroinfundibulum in intertriginous body areas including the armpits, the inguinal folds, the anogenital and inframammary regions, the perineum, and the nape [66]. It affects 2–4% of the population and it appears to be caused by an increased outer root sheath and keratinocyte proliferation in the follicular portion of the pilosebaceous unit with duct occlusion, rupture, and extrusion of its content, i.e., corneocytes, bacteria, yeast, sebum, and pilar residua, into the surrounding dermis with the development of a polymorphous inflammatory infiltrate [62]. Studies have reported an increased prevalence of MetS, dyslipidemia, and PCOS and higher levels of fasting plasma glucose, insulin, and tryglycerides in patients suffering from HS, while obesity is recognized as an exacerbating factor [67,68,69]. Moreover, HS is associated with a significantly increased risk of myocardial infarction, ischemic stroke, and cardiovascular disease-associated death [70]. Therefore, HS is now being recognized as a systemic inflammatory condition, even though the pathogenetic link with IR is still unclear; however, dysregulation of mTORC1 signaling and decreased adiponectin levels due to TNF-α overproduction have been found in patients suffering from HS [71].

4. ISAs in IR-Related Conditions and Dermatoses

The use of nutraceutical ISAs finds increasing application in the treatment of different diseases that share IR in their pathogenesis (Figure 3).

4.1. Vitamins

4.1.1. Folic Acid and Group B Vitamins

Knowles et al. and Shuster et al. highlighted for the first time the possible relationship between folic acid deficiency and skin disease in patients [72,73]. Folic acid (FoA), i.e., vitamin B9/B11, plays a crucial role in red blood cell production. In trials, supplementation with FoA reduced inflammation, OS markers, and plasma concentrations of homocysteine, and it also improved glycemic control, IR, and vitamin B12 levels in T2D [74]. Further studies highlighted that FoA intake reduced HOMA-IR and improved endothelial dysfunction in individuals with MetS and that it was beneficial for glycemic markers, including Fasting Blood Glucose (FBG) and fasting insulin [75]. However, the administration of high doses of FoA may exacerbate IR [76].
In a retrospective observational study, which enrolled 98 patients suffering from chronic plaque psoriasis and 98 healthy controls, psoriasis patients exhibited higher plasma homocysteine levels compared to controls (57% of cases vs. 25% of controls; p < 0.0001). Plasma levels of folic acid and vitamin B12 were lower in psoriatic patients, though the difference was not statistically significant. Additionally, lower levels of vitamin B12 were observed in patients with hyperhomocysteinaemia compared to those with normal homocysteine levels (p = 0.0009) [77]. Similar results were also observed in people with acne. In this context, the supplementation with folic acid and vitamin B12 during isotretinoin therapy may help prevent folate deficiency and enhance blood homocysteine levels, potentially reducing the risks of cardiovascular and neuropsychiatric disorders in patients with acne undergoing isotretinoin treatment [78].

4.1.2. Vitamin C

Besides its antioxidant properties, vitamin C (Vc) plays a role in lipid and glucose metabolism probably due to its ability to mediate adiponectin/adipoRII signaling [79]. Vc improves inflammatory conditions by reducing High-Sensitivity C-Reactive Protein (hs-CRP), interleukin (IL) 6, and FBG in obese patients with hypertension and/or diabetes, it is able to suppress visceral obesity and NAFLD, and it decreases hyperglycemia and blood pressure in T2D [80,81]. Vc is essential for maintaining skin integrity through several key mechanisms. First, it acts by modulating the keratinocyte differentiation and melanin synthesis, through the promotion and the differentiation of keratinocytes and inhibition of melanin production, contributing to a more uniform skin tone and offering protection against UV-induced photodamage. Second, it stimulates collagen synthesis, and it preserves skin barrier formation, which is vital for protecting against environmental insults and preventing dehydration. Third, as an antioxidant agent, Vc neutralizes reactive oxygen species generated by UV exposure, thereby mitigating oxidative stress and reducing the risk of photoaging and skin carcinogenesis [82]. Epidemiological studies showed that reduced plasma vitamin C levels have been observed in atopic dermatitis (AD) and porphyria cutanea tarda (PCT) patients, suggesting that deficiency may exacerbate the severity of these conditions [83,84]. While existing studies are promising, larger-scale, randomized controlled trials are necessary to confirm these findings and establish standardized treatment protocols.

4.1.3. Vitamin D

Vitamin D (Vd), i.e., cholecalciferol or 25-hydroxyvitamin D [25(OH)D], is involved in the pathogenesis of several conditions and in the modulation of lipid profiles and inflammation [85]. Studies evidenced that supplementation with Vd improves insulin sensitivity, reduces tryglycerides, low-density lipoprotein (LDL)-cholesterol, and HOMA-IR, and is beneficial in PCOS [86,87,88,89]. Further data reported that low Vd levels are associated with a greater risk of T2D, while high levels of Vd prevent T2D occurrence [90,91]. Vd influences various skin functions, including keratinocyte differentiation, immune modulation, and barrier maintenance. In addition, it regulates both innate and adaptive immune responses in the skin [92]. In psoriasis, vitamin D deficiency is commonly observed [93,94], and supplementation with vitamin D or its analogs, such as calcipotriol, has demonstrated efficacy in improving clinical symptoms by modulating immune responses, reducing keratinocyte proliferation, and inhibiting inflammation [95]. Clinical studies have shown that vitamin D treatment reduces the severity of psoriasis, particularly in combination with other therapies like narrow-band ultraviolet B (NB-UVB) phototherapy [96]. Vitamin D also modulates the Th17 cell response, which is central to psoriasis pathogenesis, and enhances the expression of antimicrobial peptides (AMPs), crucial for skin defense [97]. In AD, vitamin D deficiency is similarly associated with disease severity [98]. Vitamin D supplementation has been shown to restore immune balance by modulating Th1/Th2 cytokines and improving the epidermal barrier, thereby reducing inflammation and enhancing skin healing [99]. Clinical trials have reported improvement in AD severity upon vitamin D supplementation, particularly with respect to skin barrier integrity and immune response [100,101]. Furthermore, vitamin D’s role in modulating AMP levels, such as LL-37, is significant in combating infections common in AD, such as Staphylococcus aureus [102,103].
Despite that vitamin D supplementation shows promise in managing psoriasis and AD, the effectiveness may vary due to genetic factors and environmental conditions such as sun exposure. Further studies are required to explore the exact mechanisms of action and to optimize vitamin D-based treatments for these inflammatory skin conditions and refine treatment protocols.

4.2. Inositol

A natural isomer of glucose, inositol, is found in cell membranes as phosphatidyl-myo-inositol, the precursor of inositol triphosphate (IP3), which regulates insulin and other hormones; it is present in animal-derived food as phosphatidylinositol and in fruit and vegetables as inositol hexaphosphate (IP6) [104]. The two main inositol human isomers are D-chiro-inositol (DCI) and myo-inositol (MI) which are found in several plant-derived food, while the kidney, brain, testes, and liver can synthesize it from D-glucose at a rate of up to 4 g per day [105]. Intestinal inositol transport is driven by the sodium (Na+) gradient, and additional cofactors such as magnesium (Mg2+) increase transporter affinity (Figure 4) [106]. Pharmacokinetics investigations found that Mg2+ enhances the affinity of inositol transporter by 2.5 folds, suggesting that hypomagnesemia leads to reduced affinity of the inositol transport protein [107]. Other strategies used to improve inositol bioavailability include its association with α-lactalbumin, although it is a highly allergenic milk protein [108].
Inositol intake mitigates IR by improving lipid metabolism and reduces visceral fat, hepatic lipid accumulation, and insulin secretion [109]. Additionally, inositol increases adiponectin levels with a beneficial effect on adipogenesis, inflammation, and insulin sensitivity [110]. MI supplementation is effective in several conditions characterized by IR and MetS: trial outcomes on PCOS populations showed that MI enhanced gonadal parameters, improved glycemic and lipid profiles, ameliorated ovulation, oocyte maturation, and quality, and reduced HOMA-IR, hirsutism, and total androgen levels [111,112,113]; other studies reported that MI supplementation decreased the risk of gestational diabetes mellitus (GDM), macrosomia, and preterm birth, and it was also effective in diabetic nephropathy [114,115,116]. A recent systematic review assessed the efficacy of inositol, particularly MI and DCI, in treating various dermatological disorders. The review analyzed thirteen studies, including six randomized controlled trials, five non-randomized trials, one case series, and one case report, focusing on conditions such as acne vulgaris, hirsutism, seborrheic dermatitis, hidradenitis suppurativa, psoriasis, trichotillomania, and melanoma. Results showed that oral inositol supplementation demonstrated promising effectiveness in treating both PCOS-related and non-PCOS-related acne and hirsutism. Notably, no serious adverse effects were reported, highlighting its safety profile [117]. Currently, preparations containing MI and DCI in a 40:1 ratio are used for the treatment of PCOS with uncertain efficacy [118]. Based on this ratio the final product should include at least 300–1500 mg of DCI and 2–4 g of MI, while most available supplements provide much lower doses of DCI (i.e., 13.8–27.6 mg) [94]. Moreover, studies have shown that DCI may have a potentially harmful effect on oocytes [119]. In addition, DCI may act as a down-regulator of aromatase activity because it causes a possible increase in androgens and a decrease in estrogens, but the mechanism of action is still unknown [120].
Table 1 summarizes clinical studies conducted with oral MI in dermatological conditions.

4.3. Alpha Lipoic Acid

Alpha-lipoic acid (ALA) is a sulfur-containing fatty acid synthesized in the mitochondria, serving as a cofactor for enzymatic complexes involved in nutrient catabolism. Dietary sources include red meat, beets, carrots, potatoes, spinach, and broccoli. ALA exhibits potent antioxidant properties, neutralizing ROS through reduction of other antioxidants. Clinically, ALA is utilized in managing diabetic neuropathy by enhancing nitric oxide-mediated vasodilation and improving microcirculation [129]. It also demonstrates promise in mitigating oxidative stress-related conditions, such as ischemia–reperfusion injury and radiation-induced damage. Additionally, ALA possesses iron-chelating properties, facilitating the excretion of toxic metals like zinc, lead, and copper [130]. Being an antioxidant and an anti-inflammatory agent, ALA has been proposed recently as therapeutic option for acne vulgaris. In this regard, a preliminary study which cultured primary human sebocytes and treated them with ALA, alone or in combination with lipopolysaccharide (LPS) or dihydrotestosterone, demonstrated that ALA significantly reduced the protein expression of the inflammatory biomarkers IL-1β, IL-6, and IL-8, indicating its anti-inflammatory effect. ALA also decreased lipid peroxidation, a marker of oxidative stress [131]. In addition, ALA supplementation for age-related sensory and endothelial dysfunction in skin using two rat strains, Wistar and Brown Norway (BN), which served as models for “poorly aging” and “healthy aging”, respectively, was shown to improve vascular and sensory functions in the BN strain by the improvement of skin resistance and skin sensory thresholds and restoring the endothelial responses [132]. Therapeutic applications include ALA supplementation with dosages ranging from 600 mg to 1800 mg daily over periods up to six months. While generally safe, potential adverse effects encompass gastrointestinal disturbances, skin rashes, and hypoglycemia [133]. Contraindications involve hypersensitivity to ALA, and caution is advised in patients with thyroid disorders or those on thyroid medication. Moreover, the European Food Safety Authority (EFSA) reviewed the potential risk of insulin autoimmune syndrome (IAS) associated with the consumption of ALA. The review, based on case reports and literature reviews, concluded that ALA consumption, particularly in individuals with specific genetic polymorphisms (e.g., HLA-DRB1 * 04:03, * 04:06), increases the risk of developing IAS. This syndrome involves autoimmune-induced hypoglycemia due to insulin autoantibodies and, despite that the exact mechanism is not fully understood, it is believed that ALA may cleave insulin, altering its structure and immunogenicity, leading to antibody production. The risk of IAS was found to be relatively low in Europe but higher in countries like Japan [134].

5. Discussion

This review underscores the emerging role of IR as a key contributor to the pathogenesis and clinical course of several dermatological diseases, including psoriasis, acne vulgaris, acanthosis nigricans, and hidradenitis suppurativa. These conditions, traditionally managed as isolated cutaneous disorders, share common metabolic underpinnings driven by IR, systemic inflammation, and oxidative stress, aligning them within the broader spectrum of metabolic-associated inflammatory diseases [26,135]. The findings discussed herein suggest that ISAs, including MI, ALA, vitamin D, vitamin C, and FoA, may offer therapeutic benefits in managing these disorders by modulating glucose metabolism, attenuating oxidative stress, and restoring immune homeostasis. Among these, MI, particularly when combined with magnesium and FoA, appears to be the most extensively studied, with clinical trials demonstrating significant improvements in acne severity, hirsutism, and metabolic parameters in women with PCOS [122]. IR exacerbates skin conditions by altering glucose metabolism, inducing oxidative stress, and triggering chronic inflammation [136]. This disruption of cellular homeostasis contributes significantly to the pathogenesis of conditions like psoriasis, acne, and hidradenitis suppurativa. IR activates pathways that affect keratinocyte proliferation and differentiation, which are crucial for maintaining healthy skin structure. Therefore, targeting IR with ISAs offers a novel therapeutic approach for dermatological diseases previously considered to be primarily inflammatory or infectious in origin. In this context, ISAs are increasingly used in the treatment of dermatoses; for instance, metformin is recognized as an effective adjuvant agent in the therapeutic schemes of inflammatory dermatoses, endocrinology-related dermatosis, melasma, skin aging, and wound healing processes, and several trials have confirmed its beneficial use in acne, HS, and psoriasis, while semiglutide is effective in psoriatic patients with T2D [137,138,139,140,141]. Despite the effectiveness of conventional treatments for acne and other dermatological diseases, drugs are not without potential side effects. For instance, the use of metformin has been associated with gastrointestinal issues such as diarrhea, abdominal pain, and nausea [142]. These adverse effects can significantly reduce patient compliance and increase the likelihood of treatment failure. Additionally, long-term metformin use may lead to vitamin B12 deficiency, potentially resulting in hyperhomocysteinemia, a condition highly prevalent in dysmetabolic women [143]. In this context, preliminary data indicate that the use of nutraceutical ISAs, particularly MI, in treating IR-related skin disorders, such as acne and psoriasis, has shown positive results. MI’s ability to improve lipid metabolism, reduce visceral fat, and enhance insulin sensitivity is well-documented, particularly in conditions like PCOS [144]. Moreover, the combination of MI with other ISAs, like magnesium and folic acid, may enhance therapeutic outcomes. In this regard, the retrospective study by Pezza et al. evaluated the effects of a nutraceutical formulation containing myo-inositol, microlipodispersed magnesium, and folic acid on acne and hirsutism in women with PCOS. A total of 200 patients with acne and/or hirsutism participated and were treated for six months. The results indicated significant reductions in circulating androgen levels, including testosterone and dehydroepiandrosterone sulfate (DHEAS), along with improved insulin sensitivity, as evidenced by changes in basal insulin levels and the HOMA index. Acne severity, assessed by the global acne severity scale, and the impact on quality of life, measured by the Cardiff acne disability index and dermatology life quality index, improved substantially after three and six months of treatment. Furthermore, hirsutism, evaluated by the Ferriman–Gallwey score, also showed significant improvement. Importantly, no adverse effects were reported, ensuring high compliance [122]. Furthermore, an oral supplementation based on 2000 mg of MI, 56.25 mg of microlipodispersed Mg2+, and FoA proved to be effective in a cohort of 20 patients affected by HS, stressing the importance of an accurate assessment of metabolic profile and parameters in subjects affected by these dermatoses [128].
Several points could be addressed before ISAs can be widely implemented in clinical dermatology. First, while the agents discussed in this review are generally considered safe, there is insufficient data on their long-term safety and potential interactions with other medications commonly used in dermatology. In this regard, the effects of ISAs may vary depending on the individual’s underlying metabolic status, the severity of the dermatological condition, and genetic factors that influence insulin sensitivity. A notable limitation of the current studies is the lack of large-scale randomized controlled trials, which are essential to provide high-quality evidence on the efficacy of ISAs in managing dermatological conditions. Furthermore, the mechanisms through which ISAs exert their beneficial effects on skin health need to be better understood. While it is clear that these agents reduce inflammation and oxidative stress, the specific molecular pathways involved remain unclear.
In addition to isolated nutraceutical compounds, several botanical extracts with insulin-sensitizing properties have shown promising effects in preclinical and early clinical studies on IR-associated dermatological conditions [145]. Extracts from Berberis aristata, Cinnamomum cassia, Galega officinalis, Camellia sinensis, and Trigonella foenum-graecum have been reported to improve glycemic control, reduce systemic inflammation, and modulate adipokine levels [146,147,148]. Berberine from Berberis aristata has demonstrated the ability to activate AMPK pathways and reduce IR in both metabolic and dermatological models [149], while cinnamaldehyde from Cinnamomum cassia may influence GLUT-4 translocation and TNF-α expression [150]. Despite these encouraging findings, the clinical translation of botanical agents remains limited by significant challenges, including poor standardization of active constituents, batch-to-batch variability, and limited pharmacokinetic data [151,152]. Furthermore, the absence of regulatory harmonization and quality control among commercially available formulations complicates reproducibility and clinical applicability. Therefore, although plant-derived compounds represent a rich source of bioactive molecules with potential ISA activity, its integration into evidence-based dermatological care requires rigorous standardization, clinical validation, and toxicological assessment.
Detailed pharmacodynamic studies could provide insights into how these agents influence skin biology at a cellular level. More controlled studies are also required to determine the optimal dosages and treatment regimens for these agents in managing IR-associated dermatological diseases.
Bridging the gap between preclinical evidence and clinical implementation requires targeted efforts to address key translational challenges. Future studies should focus on translational strategies that facilitate the clinical adoption of nutraceutical ISAs in dermatology. First, biomarker-driven trials are warranted to identify subgroups of patients with insulin resistance-related dermatoses who may benefit most from ISA therapy, such as individuals with elevated HOMA-IR or altered adipokine profiles. Second, head-to-head comparisons between ISAs and conventional pharmacological agents (e.g., metformin, isotretinoin, biologics) are needed to evaluate relative efficacy, safety, and patient adherence. Third, formulation science and delivery systems (e.g., liposomal or micellar vehicles) should be explored to enhance ISA bioavailability and target skin tissues more effectively. Finally, a transdisciplinary approach involving dermatologists, endocrinologists, and clinical nutritionists will be essential for integrating ISA-based interventions into comprehensive therapeutic algorithms. The development of clinical practice guidelines incorporating nutraceuticals, once robust data from randomized controlled trials are available, will represent a key milestone in the evidence-based integration of nutraceutical ISAs into routine dermatological care.

6. Conclusions

IR is now increasingly recognized as a pathogenetic factor of several diseases as it triggers and worsens inflammation, OS, and metabolic disruption. ISAs such as MI act as metabolic regulators, antioxidants, and anti-inflammatory agents, and they are possibly effective in treating several skin disorders either when used as single agents or in association with the existing therapies. However, further clinical investigations to better assess outcomes of the use of these molecules for the treatment of dermatoses described herein are necessary.

Author Contributions

Conceptualization: A.C. and P.M.; supervision, A.C.; writing—review and editing, F.C., A.C. and A.D.P.; visualization, G.B.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data sharing is not applicable to this article.

Acknowledgments

The authors are grateful to Marta Castano for her assistance in writing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Falcetta, P.; Aragona, M.; Bertolotto, A.; Bianchi, C.; Campi, F.; Garofolo, M.; Del Prato, S. Insulin discovery: A pivotal point in medical history. Metabolism 2022, 127, 154941. [Google Scholar] [CrossRef] [PubMed]
  2. Lewis, G.F.; Brubaker, P.L. The discovery of insulin revisited: Lessons for the modern era. J. Clin. Investig. 2021, 131, e142239. [Google Scholar] [CrossRef] [PubMed]
  3. van Niekerk, G.; Christowitz, C.; Conradie, D.; Engelbrecht, A.M. Insulin as an immunomodulatory hormone. Cytokine Growth Factor Rev. 2020, 52, 34–44. [Google Scholar] [CrossRef] [PubMed]
  4. Buerger, C.; Richter, B.; Woth, K.; Salgo, R.; Malisiewicz, B.; Diehl, S.; Hardt, K.; Boehncke, S.; Boehncke, W.H. Interleukin-1Β interferes with epidermal homeostasis through induction of insulin resistance: Implications for psoriasis pathogenesis. J. Investig. Dermatol. 2012, 132, 2206–2214. [Google Scholar] [CrossRef] [PubMed]
  5. Natali, A.; Nesti, L. Vascular effects of insulin. Metabolism 2021, 124, 154891. [Google Scholar] [CrossRef]
  6. Posner, B.I. Insulin Signalling: The Inside Story. Can. J. Diabetes 2017, 41, 108–113. [Google Scholar] [CrossRef]
  7. Garcia, S.M.; Hirschberg, P.R.; Sarkar, P.; Siegel, D.M.; Teegala, S.B.; Vail, G.M.; Routh, V.H. Insulin actions on hypothalamic glucose-sensing neurones. J. Neuroendocr. 2021, 33, e12937. [Google Scholar] [CrossRef]
  8. He, Z.; Gao, Y.; Lieu, L.; Afrin, S.; Guo, H.; Williams, K.W. Acute effects of zinc and insulin on arcuate anorexigenic proopiomelanocortin neurons. Br. J. Pharmacol. 2019, 176, 725–736. [Google Scholar] [CrossRef]
  9. Norton, L.; Shannon, C.; Gastaldelli, A.; DeFronzo, R.A. Insulin: The master regulator of glucose metabolism. Metabolism 2022, 129, 155142. [Google Scholar] [CrossRef]
  10. Titchenell, P.M.; Lazar, M.A.; Birnbaum, M.J. Unraveling the Regulation of Hepatic Metabolism by Insulin. Trends Endocrinol. Metab. 2017, 28, 497–505. [Google Scholar] [CrossRef]
  11. Czech, M.P. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol. Metab. 2020, 34, 27–42. [Google Scholar] [CrossRef]
  12. Rahman, M.S.; Hossain, K.S.; Das, S.; Kundu, S.; Adegoke, E.O.; Rahman, M.A.; Hannan, M.A.; Uddin, M.J.; Pang, M.G. Role of Insulin in Health and Disease: An Update. Int. J. Mol. Sci. 2021, 22, 6403. [Google Scholar] [CrossRef]
  13. Sun, R.; Wang, J.; Li, M.; Li, J.; Pan, Y.; Liu, B.; Lip, G.Y.H.; Zhang, L. Association of Insulin Resistance with Cardiovascular Disease and All-Cause Mortality in Type 1 Diabetes: Systematic Review and Meta-analysis. Diabetes Care 2024, 47, 2266–2274. [Google Scholar] [CrossRef]
  14. Townsend, L.K.; Brunetta, H.S.; Mori, M.A.S. Mitochondria-associated ER membranes in glucose homeostasis and insulin resistance. Am. J. Physiol. Endocrinol. Metab. 2020, 319, e1053–e1060. [Google Scholar] [CrossRef] [PubMed]
  15. Yaribeygi, H.; Farrokhi, F.R.; Butler, A.E.; Sahebkar, A. Insulin resistance: Review of the underlying molecular mechanisms. J. Cell Physiol. 2019, 234, 8152–8161. [Google Scholar] [CrossRef] [PubMed]
  16. Fahed, G.; Aoun, L.; Bou Zerdan, M.; Allam, S.; Bou Zerdan, M.; Bouferraa, Y.; Assi, H.I. Metabolic Syndrome: Updates on Pathophysiology and Management in 2021. Int. J. Mol Sci. 2022, 23, 786. [Google Scholar] [CrossRef] [PubMed]
  17. Bungau, A.F.; Tit, D.M.; Stoicescu, M.; Moleriu, L.-C.; Muresan, M.; Radu, A.; Brisc, M.C.; Ghitea, T.C. Exploring a New Pathophysiological Association in Acne Vulgaris and Metabolic Syndrome: The Role of Biogenic Amines and Glutathione Peroxidase. Medicinal 2024, 60, 513. [Google Scholar] [CrossRef]
  18. Qu, X.; Donnelly, R. Sex Hormone-Binding Globulin (SHBG) as an Early Biomarker and Therapeutic Target in Polycystic Ovary Syndrome. Int. J. Mol. Sci. 2020, 21, 8191. [Google Scholar] [CrossRef]
  19. Armanini, D.; Boscaro, M.; Bordin, L.; Sabbadin, C. Controversies in the Pathogenesis, Diagnosis and Treatment of PCOS: Focus on Insulin Resistance, Inflammation, and Hyperandrogenism. Int. J. Mol. Sci. 2022, 23, 4110. [Google Scholar] [CrossRef]
  20. da Silva Rosa, S.C.; Nayak, N.; Caymo, A.M.; Gordon, J.W. Mechanisms of muscle insulin resistance and the cross-talk with liver and adipose tissue. Physiol. Rep. 2020, 8, e14607. [Google Scholar] [CrossRef]
  21. Goh, L.P.W.; Sani, S.A.; Sabullah, M.K.; Gansau, J.A. The Prevalence of Insulin Resistance in Malaysia and Indonesia: An Updated Systematic Review and Meta-Analysis. Medicinal 2022, 58, 826. [Google Scholar] [CrossRef]
  22. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef]
  23. Giammò, A.; Sarzi-Puttini, P.; Favro, M.; De Pedrini, A.; Baj, G. Rationale of Insulin-Sensitizing Agents in the Treatment of Functional Bladder Disorders. J. Biol. Regul. Homeost. Agents 2023, 37, 4499–4510. [Google Scholar]
  24. Dainichi, T.; Iwata, M. Inflammatory loops in the epithelial-immune microenvironment of the skin and skin appendages in chronic inflammatory diseases. Front. Immunol. 2023, 14, 1274270. [Google Scholar] [CrossRef] [PubMed]
  25. Napolitano, M.; Megna, M.; Monfrecola, G. Insulin resistance and skin diseases. Sci. World J. 2015, 2015, 479354. [Google Scholar] [CrossRef] [PubMed]
  26. Hu, Y.; Zhu, Y.; Lian, N.; Chen, M.; Bartke, A.; Yuan, R. Metabolic Syndrome and Skin Diseases. Front. Endocrinol. 2019, 10, 788. [Google Scholar] [CrossRef] [PubMed]
  27. Alajroush, W.A.; Alrshid, A.I.; Alajlan, A.H.; Alsalamah, Y.B.; Alhumaidan, M.I.; Alhoumedan, A.I.; Alrasheed, M.I.; Alowairdhi, Y.A.; Alowirdi, F.; Aljoufi, A.Z.; et al. Psoriasis and Metabolic Disorders: A Comprehensive Meta-Analysis of Million Adults Worldwide. Cereus 2024, 16, e52099. [Google Scholar] [CrossRef]
  28. Armstrong, A.W.; Read, C. Pathophysiology, Clinical Presentation, and Treatment of Psoriasis: A Review. JAMA 2020, 323, 1945–1960. [Google Scholar] [CrossRef]
  29. Dinić, M.; Zečević, R.D.; Hajduković, Z.; Mijušković, M.; Đurić, P.; Jović, Z.; Grdinić, A.; Petrović, M.; Terzić, B.; Pejović, J.; et al. Psoriasis is the independent factor for early atherosclerosis: A prospective study of cardiometabolic risk profile. Vojn. Pregl. 2016, 73, 1094–1101. [Google Scholar] [CrossRef]
  30. Chiu, H.Y.; Hung, C.J.; Muo, C.H.; Fan, K.C.; Sung, F.C. The bidirectional association between type 2 diabetes and psoriasis: Two retrospective cohort studies. Indian J. Dermatol. Venereol. Leprol. 2020, 86, 366–374. [Google Scholar]
  31. Eder, L.; Chandran, V.; Cook, R.; Gladman, D.D. The risk of developing diabetes mellitus in patients with psoriatic arthritis: A cohort study. J. Rheumatol. 2017, 44, 286–291. [Google Scholar] [CrossRef]
  32. Babalic, F.C.A.; Borza, C.; Ilie Rosca, C.; Gurban, C.V.; Banciu, C.D.; Mederle, O.A.; Popa, M.D.; Chelu, S.C.; Marius, P.; Sharma, A.; et al. Endothelial dysfunction in psoriatic arthritis patients: Correlations between insulin resistance and disease activity. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 6796–6804. [Google Scholar]
  33. Wang, F.; Wang, Y.; Kong, X.; Mu, J.; Wang, Z.; Yang, X.; Ye, J. Association between psoriasis and serum apolipoprotein A1 and B: A systematic review and meta-analysis. Heliyon 2023, 9, e21168. [Google Scholar] [CrossRef] [PubMed]
  34. Słuczanowska-Głabowska, S. Adiponectin, Leptin and Resistin in Patients with Psoriasis. J. Clin. Med. 2023, 12, 663. [Google Scholar] [CrossRef] [PubMed]
  35. Kiełbowski, K.; Bakinowska, E.; Ostrowski, P.; Pala, B.; Gromowska, E.; Gurazda, K.; Dec, P.; Modrzejewski, A.; Pawlik, A. The Role of Adipokines in the Pathogenesis of Psoriasis. Int. J. Mol. Sci. 2023, 24, 6390. [Google Scholar] [CrossRef] [PubMed]
  36. Furue, K. Psoriasis and the TNF/IL23/IL17 axis. G. Ital. Dermatol. Venereol. 2019, 154, 418–424. [Google Scholar] [CrossRef]
  37. Sarandi, E.; Krueger-Krasagakis, S.; Tsoukalas, D.; Sidiropoulou, P.; Evangelou, G.; Sifaki, M.; Rudofsky, G.; Drakoulis, N.; Tsatsakis, A. Psoriasis immunometabolism: Progress on metabolic biomarkers and targeted therapy. Front. Mol. Biosci. 2023, 10, 1201912. [Google Scholar] [CrossRef]
  38. Polic, M.V.; Miskulin, M.; Smolic, M.; Kralik, K.; Miskulin, I.; Berkovic, M.C.; Curcic, I.B. Psoriasis Severity-A Risk Factor of Insulin Resistance Independent of Metabolic Syndrome. Int. J. Environ. Res. Public Health 2018, 15, 1486. [Google Scholar] [CrossRef]
  39. Su, R. Metabolic influences on T cell in psoriasis: A literature review. Front. Immunol. 2023, 14, 1279846. [Google Scholar] [CrossRef]
  40. Lee, T.H.; Wu, C.H.; Chen, M.L.; Yip, H.T.; Lee, C.I.; Lee, M.S.; Wei, J.C. Risk of Psoriasis in Patients with Polycystic Ovary Syndrome: A National Population-Based Cohort Study. J. Clin. Med. 2020, 9, 1947. [Google Scholar] [CrossRef]
  41. Jeanes, Y.M.; Reeves, S. Metabolic consequences of obesity and insulin resistance in polycystic ovary syndrome: Diagnostic and methodological challenges. Nutr. Res. Rev. 2017, 30, 97–105. [Google Scholar] [CrossRef]
  42. Melnik, B.C. Acne vulgaris: The metabolic syndrome of the pilosebaceous follicle. Clin. Dermatol. 2018, 36, 29–40. [Google Scholar] [CrossRef]
  43. Tan, J.K.L.; Stein Gold, L.F.; Alexis, A.F.; Harper, J.C. Current Concepts in Acne Pathogenesis: Pathways to Inflammation. Semin. Cutan. Med. Surg. 2018, 37, S60–S62. [Google Scholar] [CrossRef]
  44. Sadowska-Przytocka, A.; Gruszczyńska, M.; Ostałowska, A.; Antosik, P.; Czarnecka-Operacz, M.; Adamski, Z.; Łącka, K. Insulin resistance in the course of acne—Literature review. Postepy. Dermatol. Alergol. 2022, 39, 231–238. [Google Scholar] [CrossRef] [PubMed]
  45. Clayton, R.W.; Göbel, K.; Niessen, C.M.; Paus, R.; van Steensel, M.A.M.; Lim, X. Homeostasis of the sebaceous gland and mechanisms of acne pathogenesis. Br. J. Dermatol. 2019, 181, 677–690. [Google Scholar] [CrossRef] [PubMed]
  46. Langan, E.A.; Hinde, E.; Paus, R. Prolactin as a candidate sebotrop(h)ic hormone? Exp. Dermatol. 2018, 27, 729–736. [Google Scholar] [CrossRef] [PubMed]
  47. Çerman, A.A.; Aktaş, E.; Altunay, İ.K.; Arıcı, J.E.; Tulunay, A.; Ozturk, F.Y. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. J. Am. Acad. Dermatol. 2016, 75, 155–162. [Google Scholar] [CrossRef]
  48. Stewart, T.J.; Bazergy, C. Hormonal and dietary factors in acne vulgaris versus controls. Derm.-Endocrinol. 2018, 10, e1442160. [Google Scholar] [CrossRef]
  49. Snast, I.; Dalal, A.; Twig, G.; Astman, N.; Kedem, R.; Levin, D.; Erlich, Y.; Leshem, Y.A.; Lapidoth, M.; Hodak, E.; et al. Acne and obesity: A nationwide study of 600,404 adolescents. J. Am. Acad. Dermatol. 2019, 81, 723–729. [Google Scholar] [CrossRef]
  50. Balta, I.; Ekiz, O.; Ozuguz, P.; Ustun, I.; Karaca, S.; Dogruk Kacar, S.; Eksioglu, M. Insulin resistance in patients with post-adolescent acne. Int. J. Dermatol. 2015, 54, 662–666. [Google Scholar] [CrossRef]
  51. Kartal, D.; Yildiz, H.; Ertas, R.; Borlu, M.; Utas, S. Association between isolated female acne and insulin resistance: A prospective study. G Ital. Dermatol. Venereol. 2016, 151, 353–357. [Google Scholar]
  52. Emiroğlu, N.; Cengiz, F.P.; Kemeriz, F. Insulin resistance in severe acne vulgaris. Adv. Dermatol. Allergol. 2015, 32, 281–285. [Google Scholar] [CrossRef]
  53. Bungau, A.F.; Radu, A.F.; Bungau, S.G.; Vesa, C.M.; Tit, D.M.; Endres, L.M. Oxidative stress and metabolic syndrome in acne vulgaris: Pathogenetic connections and potential role of dietary supplements and phytochemicals. Biomed. Pharmacother. 2023, 164, 115003. [Google Scholar] [CrossRef]
  54. Liu, H.-M.; Cheng, M.-Y.; Xun, M.-H.; Zhao, Z.-W.; Zhang, Y.; Tang, W.; Cheng, J.; Ni, J.; Wang, W. Possible Mechanisms of Oxidative Stress-Induced Skin Cellular Senescence, Inflammation, and Cancer and the Therapeutic Potential of Plant Polyphenols. Int. J. Mol. Sci. 2023, 24, 3755. [Google Scholar] [CrossRef]
  55. Dreno, B.; Dagnelie, M.A.; Khammari, A.; Corvec, S. The Skin Microbiome: A New Actor in Inflammatory Acne. Am. J. Clin. Dermatol. 2020, 21, 18–24. [Google Scholar] [CrossRef]
  56. Das, A.; Datta, D.; Kassir, M.; Wollina, U.; Galadari, H.; Lotti, T.; Jafferany, M.; Grabbe, S.; Goldust, M. Acanthosis nigricans: A review. J. Cosmet. Dermatol. 2020, 19, 1857–1865. [Google Scholar] [CrossRef] [PubMed]
  57. Leung, A.K.C.; Lam, J.M.; Barankin, B.; Leong, K.F.; Hon, K.L. Acanthosis Nigricans: An Updated Review. Curr. Pediatr. Rev. 2022, 19, 68–82. [Google Scholar] [CrossRef] [PubMed]
  58. Ng, H.Y. Acanthosis nigricans in obese adolescents: Prevalence, impact, and management challenges. Adolesc. Health Med. Ther. 2016, 8, 1–10. [Google Scholar] [CrossRef] [PubMed]
  59. Banti, S.; Sumathy, T.K.; Pramila, K. Insulin resistance in various grades of acanthosis nigricans. Acta Dermatovenerol. Alp. Pannonica Adriat. 2022, 31, 101–104. [Google Scholar] [CrossRef]
  60. Radu, A.M.; Carsote, M.; Dumitrascu, M.C.; Sandru, F. Acanthosis Nigricans: Pointer of Endocrine Entities. Diagnostics 2022, 12, 2519. [Google Scholar] [CrossRef]
  61. Philip, N.E.; Girisha, B.S.; Shetty, S.; Pinto, A.M.; Noronha, T.M. Estimation of Metabolic Syndrome in Acanthosis Nigricans—A Hospital Based Cross-Sectional Study. Indian J. Dermatol. 2022, 67, 92. [Google Scholar] [CrossRef]
  62. Velazquez-Bautista, M.; López-Sandoval, J.J.; González-Hita, M.; Vázquez-Valls, E.; Cabrera-Valencia, I.Z.; Torres-Mendoza, B.M. Association of metabolic syndrome with low birth weight, intake of high-calorie diets and acanthosis nigricans in children and adolescents with overweight and obesity. Endocrinol. Diabetes Nutr. 2017, 64, 11–17. [Google Scholar] [CrossRef]
  63. Ali, S.Y.; Manne, V.; Manne, R.; Himani, C. Neurofibromatosis, Down’s syndrome, and acquired abnormalities. Indian Dermatol. Online J. 2016, 7, 198–200. [Google Scholar] [CrossRef]
  64. O’Brien, B.; Dahiya, R.; Kimble, R. Hyperandrogenism, insulin resistance and acanthosis nigricans (HAIR-AN syndrome): An extreme subphenotype of polycystic ovary syndrome. BMJ Case Rep. 2020, 13, e231749. [Google Scholar] [CrossRef] [PubMed]
  65. Maguolo, A.; Maffeis, C. Acanthosis nigricans in childhood: A cutaneous marker that should not be underestimated, especially in obese children. Acta Paediatr. 2020, 109, 481–487. [Google Scholar] [CrossRef] [PubMed]
  66. Abu Rached, N.; Gambichler, T.; Dietrich, J.W.; Ocker, L.; Seifert, C.; Stockfleth, E.; Bechara, F.G. The Role of Hormones in Hidradenitis Suppurativa: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 15250. [Google Scholar] [CrossRef] [PubMed]
  67. Özkur, E.; Erdem, Y.; Altunay, İ.K.; Demir, D.; Dolu, N.Ç.; Serin, E.; Çerman, A.A. Serum irisin level, insulin resistance, and lipid profiles in patients with hidradenitis suppurativa: A case-control study. An. Bras. Dermatol. 2020, 95, 708–713. [Google Scholar] [CrossRef]
  68. Shen, A.S.; Johnson, J.S.; Kerns, M.L. Dietary Factors and Hidradenitis Suppurativa. Dermatol. Ther. 2023, 13, 3007–3017. [Google Scholar] [CrossRef]
  69. Mintoff, D.; Agius, R.; Fava, S.; Pace, N.P. Investigating Adiposity-Related Metabolic Health Phenotypes in Patients with Hidradenitis Suppurativa: A Cross-Sectional Study. J. Clin. Med. 2023, 12, 4847. [Google Scholar] [CrossRef]
  70. Hambly, R.; Kearney, N.; Hughes, R.; Fletcher, J.M.; Kirby, B. Metformin Treatment of Hidradenitis Suppurativa: Effect on Metabolic Parameters, Inflammation, Cardiovascular Risk Biomarkers, and Immune Mediators. Int. J. Mol. Sci. 2023, 24, 6969. [Google Scholar] [CrossRef]
  71. Zouboulis, C.C.; Benhadou, F.; Byrd, A.S.; Chandran, N.S.; Giamarellos-Bourboulis, E.J.; Fabbrocini, G. What causes hidradenitis suppurativa?-15 years after. Exp. Dermatol. 2020, 29, 1154–1170. [Google Scholar] [CrossRef] [PubMed]
  72. Knowles, J.P.; Shuster, S.; Wells, G.C. Folic-acid deficiency in patients with skin disease. Lancet 1963, 1, 1138–1139. [Google Scholar] [CrossRef] [PubMed]
  73. Shuster, S.; Marks, J.; Chanarin, I. Folic acid deficiency in patients with skin disease. Br. J. Dermatol. 1967, 79, 398–402. [Google Scholar] [CrossRef] [PubMed]
  74. Asbaghi, O.; Ashtary-Larky, D.; Bagheri, R.; Moosavian, S.P.; Olyaei, H.P.; Nazarian, B.; Rezaei Kelishadi, M.; Wong, A.; Candow, D.G.; Dutheil, F.; et al. Folic Acid Supplementation Improves Glycemic Control for Diabetes Prevention and Management: A Systematic Review and Dose-Response Meta-Analysis of Randomized Controlled Trials. Nutrients 2021, 13, 2355. [Google Scholar] [CrossRef]
  75. Setola, E.; Monti, L.D.; Galluccio, E.; Palloshi, A.; Fragasso, G.; Paroni, R.; Magni, F.; Sandoli, E.P.; Lucotti, P.; Costa, S.; et al. Insulin resistance and endothelial function are improved after folate and vitamin B12 therapy in patients with metabolic syndrome: Relationship between homocysteine levels and hyperinsulinemia. Eur. J. Endocrinol. 2004, 151, 483–489. [Google Scholar] [CrossRef]
  76. Karaçil Ermumcu, M.Ş.; Acar Tek, N. Effects of High-dose Folic Acid Supplementation on Maternal/Child Health Outcomes: Gestational Diabetes Mellitus in Pregnancy and Insulin Resistance in Offspring. Can. J. Diabetes 2023, 47, 133–142. [Google Scholar] [CrossRef]
  77. Brazzelli, V.; Grasso, V.; Fornara, L.; Moggio, E.; Gamba, G.; Villani, S.; Borroni, G. Homocysteine, vitamin B12 and folic acid levels in psoriatic patients and correlation with disease severity. Int. J. Immunopathol. Pharmacol. 2010, 23, 911–916. [Google Scholar] [CrossRef]
  78. Ghiasi, M.; Mortazavi, H.; Jafari, M. Efficacy of Folic Acid and Vitamin B12 Replacement Therapies in the Reduction of Adverse Effects of Isotretinoin: A Randomized Controlled Trial. Skinmed 2018, 16, 239–245. [Google Scholar]
  79. Villagran, M.; Ferreira, J.; Martorell, M.; Mardones, L. The Role of Vitamin C in Cancer Prevention and Therapy: A Literature Review. Antioxidants 2021, 10, 1894. [Google Scholar] [CrossRef]
  80. Ellulu, M.S.; Rahmat, A.; Patimah, I.; Khaza’ai, H.; Abed, Y. Effect of vitamin C on inflammation and metabolic markers in hypertensive and/or diabetic obese adults: A randomized controlled trial. Drug. Des. Devel. Ther. 2015, 9, 3405–3412. [Google Scholar] [CrossRef]
  81. Lee, H.; Ahn, J.; Shin, S.S.; Yoon, M. Ascorbic acid inhibits visceral obesity and nonalcoholic fatty liver disease by activating peroxisome proliferator-activated receptor α in high-fat-diet fed C57BL/6J mice. Int. J. Obes. 2019, 43, 1620–1630. [Google Scholar] [CrossRef]
  82. Wang, K.; Jiang, H.; Li, W.; Qiang, M.; Dong, T.; Li, H. Role of Vitamin C in Skin Diseases. Front. Physiol. 2018, 9, 819. [Google Scholar] [CrossRef]
  83. Anderson, K.E. Porphyria cutanea tarda: A possible role for ascorbic acid. Hepatology 2007, 45, 6–8. [Google Scholar] [CrossRef] [PubMed]
  84. Sinclair, P.R.; Gorman, N.; Shedlofsky, S.I.; Honsinger, C.P.; Sinclair, J.F.; Karagas, M.R.; Anderson, K.E. Ascorbic acid deficiency in porphyria cutanea tarda. J. Lab Clin. Med. 1997, 130, 197–201. [Google Scholar] [CrossRef] [PubMed]
  85. Maretzke, F.; Bechthold, A.; Egert, S.; Ernst, J.B.; Melo van Lent, D.; Pilz, S.; Reichrath, J.; Stangl, G.I.; Stehle, P.; Volkert, D.; et al. Role of Vitamin D in Preventing and Treating Se- 12 lected Extraskeletal Diseases-An Umbrella Review. Nutrients 2020, 12, 969. [Google Scholar] [CrossRef] [PubMed]
  86. Cefalo, C.M.A.; Conte, C.; Sorice, G.P.; Moffa, S.; Sun, V.A.; Cinti, F.; Salomone, E.; Muscogiuri, G.; Brocchi, A.A.G.; Pontecorvi, A.; et al. Effect of Vitamin D Supplementation on Obesity-Induced Insulin Resistance: A Double-Blind, Randomized, Placebo-Controlled Trial. Obesity 2018, 26, 651–657. [Google Scholar] [CrossRef]
  87. Jamilian, M.; Samimi, M.; Ebrahimi, F.A.; Hashemi, T.; Taghizadeh, M.; Razavi, M. The effects of vitamin D and omega-3 fatty acid co-supplementation on glycemic control and lipid concentrations in patients with gestational diabetes. J. Clin. Lipidol. 2017, 11, 459–468. [Google Scholar] [CrossRef]
  88. Imga, N.N.; Karci, A.C.; Oztas, D.; Berker, D.; Guler, S. Effects of vitamin D supplementation on insulin resistance and dyslipidemia in overweight and obese premenopausal women. AMS 2019, 15, 598–606. [Google Scholar] [CrossRef]
  89. Łagowska, K.; Bajerska, J.; Jamka, M. The Role of Vitamin D Oral Supplementation in Insulin Resistance in Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2018, 10, 1637. [Google Scholar] [CrossRef]
  90. Afzal, S.; Bojesen, S.E.; Nordestgaard, B.G. Low 25- hydroxyvitamin D and risk of type 2 diabetes: A prospective cohort study and metaanalysis. Clin. Chem. 2013, 59, 381–391. [Google Scholar] [CrossRef]
  91. Song, Y.; Wang, L.; Pittas, A.G.; Del Gobbo, L.; Zhang, C.; Manson, J.E.; Hu, F.B. Blood 25-hydroxy vitamin D levels and incident type 2 diabetes: A meta-analysis of prospective studies. Diabetes Care 2013, 36, 1422–1428. [Google Scholar] [CrossRef]
  92. Bikle, D.D. Vitamin D and the skin: Physiology and pathophysiology. Rev. Endocr. Metab. Disord. 2012, 13, 3–19. [Google Scholar] [CrossRef] [PubMed]
  93. Gisondi, P.; Rossini, M.; Di Cesare, A.; Idolazzi, L.; Farina, S.; Beltrami, G.; Peris, K.; Girolomoni, G. Vitamin D status in patients with chronic plaque psoriasis. Br. J. Dermatol. 2012, 166, 505–510. [Google Scholar] [CrossRef]
  94. Wilson, P.B. Serum 25-hydroxyvitamin D status in individuals with psoriasis in the general population. Endocrine 2013, 44, 537–539. [Google Scholar] [CrossRef]
  95. Finamor, D.C.; Sinigaglia-Coimbra, R.; Neves, L.C.; Gutierrez, M.; Silva, J.J.; Torres, L.D.; Surano, F.; Neto, D.J.; Novo, N.F.; Juliano, Y.; et al. A pilot study assessing the effect of prolonged administration of high daily doses of vitamin D on the clinical course of vitiligo and psoriasis. J. Am. Acad. Dermatol. 2013, 68, 511–512. [Google Scholar] [CrossRef]
  96. Al-Mutairi, N.; Shaaban, D. Effect of narrow-band ultraviolet B therapy on serum vitamin D and cathelicidin (LL-37) in patients with chronic plaque psoriasis. J. Cutan. Med. Surg. 2014, 18, 43–48. [Google Scholar] [CrossRef]
  97. Mattozzi, C.; Paolino, G.; Salvi, M.; Macaluso, L.; Luci, C.; Morrone, S.; Calvieri, S.; Richetta, A.G. Importance of regulatory T cells in the pathogenesis of psoriasis: Review of the literature. Dermatology 2013, 227, 134–145. [Google Scholar] [CrossRef]
  98. Dall’Ara, F.; Riccio, L.; Santoro, A.; Di Mattia, D.; Di Pietro, C.; Chiricozzi, A.; Calzavara-Pinton, P.; Girolomoni, G. The association between serum vitamin D levels and the severity of atopic dermatitis in children and adults: A systematic review. Dermatitis 2014, 25, 1–11. [Google Scholar]
  99. Ooi, H.S.; Kamal, S.M.; Yeo, J.F.; Thng, T.G.S.; Giam, Y.C. A systematic review of the effect of vitamin D supplementation on atopic dermatitis. Dermatology 2017, 233, 356–361. [Google Scholar]
  100. Gilaberte, Y.; Rojas-Hernandez, C.; Ramos, L.; Granizo, C.; Bardají, M.; Herranz, P.; Burgos, F.J. Efficacy of vitamin D supplementation in atopic dermatitis patients with low vitamin D levels: Results of a randomized controlled trial. Eur. J. Dermatol. 2017, 27, 372–377. [Google Scholar]
  101. Drozdenko, G.; Heine, G.; Worm, M. Oral vitamin D increases the frequencies of CD38+ human B cells and ameliorates IL-17-producing T cells. Exp Dermatol. 2014, 23, 107–112. [Google Scholar] [CrossRef] [PubMed]
  102. Kanda, N.; Hau, C.S.; Tada, Y.; Sato, S.; Watanabe, S. Decreased serum LL-37 and vitamin D3 levels in atopic dermatitis: Relationship between IL-31 and oncostatin M. Allergy 2012, 67, 804–812. [Google Scholar] [CrossRef] [PubMed]
  103. Gilaberte, Y.; Sanmartín, R.; Aspiroz, C.; Hernandez-Martin, A.; Benito, D.; Sanz-Puertolas, P.; Alonso, M.; Torrelo, A.; Torres, C. Correlation Between Serum 25-Hydroxyvitamin D and Virulence Genes of Staphylococcus aureus Isolates Colonizing Children with Atopic Dermatitis. Pediatr Dermatol. 2015, 32, 506–513. [Google Scholar] [CrossRef] [PubMed]
  104. Di Nicolantonio, J.J.; O’Keefe, J. Myo-inositol for insulin resistance, metabolic syndrome, polycystic ovary syndrome and gestational diabetes. Open Heart 2022, 9, e001989. [Google Scholar] [CrossRef]
  105. Antonowski, T.; Osowski, A.; Lahuta, L.; Górecki, R.; Rynkiewicz, A.; Wojtkiewicz, J. Health-Promoting Properties of Selected Cyclitols for Metabolic Syndrome and Diabetes. Nutrients 2019, 11, 2314. [Google Scholar] [CrossRef]
  106. López-Gambero, A.J.; Sanjuan, C.; Serrano-Castro, P.J.; Suárez, J.; Rodríguez de Fonseca, F. The Biomedical Uses of Inositols: A Nutraceutical Approach to Metabolic Dysfunction in Aging and Neurodegenerative Diseases. Biomedicines 2020, 8, 295. [Google Scholar] [CrossRef]
  107. Grafton, G.; Bunce, C.M.; Sheppard, M.C.; Brown, G.; Baxter, M.A. Effect of Mg2+ on Na(+)-dependent inositol transport. Role for Mg2+ in etiology of diabetic complications. Diabetes 1992, 41, 35–39. [Google Scholar] [CrossRef]
  108. Kamenov, Z.; Gateva, A.; Dinicola, S.; Unfer, V. Comparing the Efficacy of Myo-Inositol Plus α-Lactalbumin vs. Myo-Inositol Alone on Reproductive and Metabolic Disturbances of Polycystic Ovary Syndrome. Metabolites 2023, 13, 717. [Google Scholar] [CrossRef]
  109. Zarezadeh, M.; Dehghani, A.; Faghfouri, A.H.; Radkhah, N.; Naemi Kermanshahi, M.; Hamedi Kalajahi, F.; Mohammadzadeh Honarvar, N.; Ghoreishi, Z.; Ostadrahimi, A.; Ebrahimi Mamaghani, M. Inositol supplementation and body mass index: A systematic review and meta-analysis of randomized clinical trials. Obes. Sci. Pract. 2021, 8, 387–397. [Google Scholar] [CrossRef]
  110. Alexandraki, K.I.; Kandaraki, E.A.; Poulia, K.A.; Piperi, C.; Papadimitriou, E.; Papaioannou, T.G. Assessment of Early Markers of Cardiovascular Risk in Polycystic Ovary Syndrome. Touch Rev. Endocrinol. 2021, 17, 37–53. [Google Scholar]
  111. Genazzani, A.D.; Lanzoni, C.; Ricchieri, F.; Jasonni, V.M. Myoinositol administration positively affects hyperinsulinemia and hormonal parameters in overweight patients with polycystic ovary syndrome. Gynecol. Endocrinol. 2008, 24, 139–144. [Google Scholar] [CrossRef] [PubMed]
  112. Shokrpour, M.; Foroozanfard, F.; Afshar Ebrahimi, F.; Vahedpoor, Z.; Aghadavod, E.; Ghaderi, A.; Asemi, Z. Comparison of myo-inositol and metformin on glycemic control, lipid profiles, and gene expression related to insulin and lipid metabolism in women with polycystic ovary syndrome: A randomized controlled clinical trial. Gynecol. Endocrinol. 2019, 35, 406–411. [Google Scholar] [CrossRef] [PubMed]
  113. Minozzi, M.; Costantino, D.; Guaraldi, C.; Unfer, V. The effect of a combination therapy with myo-inositol and a combined oral contraceptive pill versus a combined oral contraceptive pill alone on metabolic, endocrine, and clinical parameters in polycystic ovary syndrome. Gynecol. Endocrinol. 2011, 27, 920–924. [Google Scholar] [CrossRef]
  114. Regidor, P.A.; Schindler, A.E. Myoinositol as a Safe and Alternative Approach in the Treatment of Infertile PCOS Women: A German Observational Study. Int. J. Endocrinol. 2016, 2016, 1–5. [Google Scholar] [CrossRef]
  115. Santamaria, A.; Alibrandi, A.; Di Benedetto, A.; Pintaudi, B.; Corrado, F.; Facchinetti, F.; D’Anna, R. Clinical and metabolic outcomes in pregnant women at risk for gestational diabetes mellitus supplemented with myo-inositol: A secondary analysis from 3 RCTs. Am. J. Obstet. Gynecol. 2018, 219, 300.e1–300.e6. [Google Scholar] [CrossRef]
  116. Nayak, B.; Kondeti, V.K.; Xie, P.; Lin, S.; Viswakarma, N.; Raparia, K.; Kanwar, Y.S. Transcriptional and post-translational modulation of myo-inositol oxygenase by high glucose and related pathobiological stresses. J. Biol. Chemi. 2011, 286, 27594–27611. [Google Scholar] [CrossRef]
  117. Quach, M.; Chang, Y.F.; Lee, I.; Tai, L.; Choi, F.; Bodemer, A. Inositol for Treating Dermatological Disorders: A Systematic Review. J. Integr. Dermatol. 2024. Available online: https://www.jintegrativederm.org/article/122716 (accessed on 18 May 2025).
  118. Salomone, S.; Carruba, M.; Drago, F. Gli Inositoli Nella sindrome Dell’ovaio Policistico: Evidenze Cliniche e Razionali D’impiego; Opinion Papers; Italian Society of Pharmacology: Milan, Italy, 2017; pp. 1–22. Available online: https://www.sifweb.org/pubblicazioni/position-papers/position-papers-gli-inositoli-nella-sindrome-dell-ovaio-policistico-evidenze-cliniche-e-razionale-d-impiego-2017-03-01 (accessed on 18 May 2025).
  119. Bevilacqua, A.; Dragotto, J.; Lucarelli, M.; Di Emidio, G.; Monastra, G.; Tatone, C. High Doses of D-Chiro-Inositol Alone Induce a PCO-Like Syndrome and Other Alterations in Mouse Ovaries. Int. J. Mol. Sci. 2021, 22, 5691. [Google Scholar] [CrossRef]
  120. Monastra, G.; Vazquez-Levin, M.; Bezerra Espinola, M.S.; Bilotta, G.; Laganà, A.S.; Unfer, V. D-chiro-inositol, an aromatase down-modulator, increases androgens and reduces estrogens in male volunteers: A pilot study. Basic Clin. Androl. 2021, 31, 13. [Google Scholar] [CrossRef]
  121. Pezza, M.; Carlomagno, V.; Casucci, G. Inositol and acne. G Ital. Dermatol. Venereol. 2015, 150, 649–653. [Google Scholar]
  122. Pezza, M.; Carlomagno, V.; Sammarco, E.; Trischitta, A.; Ceddia, C.; Vitiello, A.; Baj, G.; Citi, V.; Colletti, A. Association of Myo-Inositol and Microlipodispersed Magnesium in Androgen-Dependent Dermatological Diseases: A Retrospective Study. Pharmaceuticals 2025, 18, 251. [Google Scholar] [CrossRef]
  123. Fruzzetti, F.; Perini, D.; Russo, M.; Bucci, F.; Gadducci, A. Comparison of two insulin sensitizers, metformin and myo-inositol, in women with polycystic ovary syndrome (PCOS). Gynecol. Endocrinol. 2016, 33, 39–42. [Google Scholar] [CrossRef] [PubMed]
  124. Minozzi, M.; D’Andrea, G.; Unfer, V. Treatment of hirsutism with myo-inositol: A prospective clinical study. Reprod. Biomed. Online 2008, 17, 579–582. [Google Scholar] [CrossRef] [PubMed]
  125. Advani, K.; Batra, M.; Tajpuriya, S.; Gupta, R.; Saraswat, A.; Nagar, H.D.; Makwana, L.; Kshirsagar, S.; Kaul, P.; Ghosh, A.K.; et al. Efficacy of combination therapy of inositols, antioxidants and vitamins in obese and non-obese women with polycystic ovary syndrome: An observational study. J. Obstet. Gynaecol. 2020, 40, 96–101. [Google Scholar] [CrossRef] [PubMed]
  126. Ramanan, E.A.; Ravi, S.; Anbu, K.R.R.; Michael, M. Efficacy and Safety of Tracnil™ Administration in Patients with Dermatological Manifestations of PCOS: An Open-Label Single-Arm Study. Dermatol. Res. Pract. 2020, 2020, 1–10. [Google Scholar] [CrossRef]
  127. Allan, S.; Kavanagh, G.; Herd, R.; Savin, J. The effect of inositol supplements on the psoriasis of patients taking lithium: A randomized, placebo-controlled trial. Br. J. Dermatol. 2004, 150, 966–969. [Google Scholar] [CrossRef]
  128. Donnarumma, M.; Marasca, C.; Palma, M.; Vastarella, M.; Annunziata, M.C.; Fabbrocini, G. An oral supplementation based on myo-inositol, folic acid and liposomal magnesium may act synergistically with antibiotic therapy and can improve metabolic profile in patients affected by Hidradenitis suppurativa: Our experience. G Ital. Dermatol. Venereol. 2020, 155, 749–753. [Google Scholar] [CrossRef]
  129. Abubaker, S.A.; Alonazy, A.M.; Abdulrahman, A. Effect of Alpha-Lipoic Acid in the Treatment of Diabetic Neuropathy: A Systematic Review. Cureus 2022, 14, e25750. [Google Scholar] [CrossRef]
  130. Shay, K.P.; Moreau, R.F.; Smith, E.J.; Smith, A.R.; Hagen, T.M. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim. Biophys. Acta 2009, 1790, 1149–1160. [Google Scholar] [CrossRef]
  131. Lee, W.J.; Eun, D.H.; Kim, S.M.; Kim, J.Y.; Jang, Y.H.; Lee, S.J. Anti-Inflammatory and Antioxidative Effects of Alpha Lipoic Acid on Cultured Human Sebocytes. Ann. Dermatol. 2019, 31, 84–87. [Google Scholar] [CrossRef]
  132. de Bengy, A.-F.; Decorps, J.; Martin, L.S.; Pagnon, A.; Chevalier, F.P.; Sigaudo-Roussel, D.; Fromy, B. Alpha-Lipoic Acid Supplementation Restores Early Age-Related Sensory and Endothelial Dysfunction in the Skin. Biomedicines 2022, 10, 2887. [Google Scholar] [CrossRef]
  133. Jibril, A.T.; Jayedi, A.; Shab-Bidar, S. Efficacy and safety of oral alpha-lipoic acid supplementation for type 2 diabetes management: A systematic review and dose-response meta-analysis of randomized trials. Endocr. Connect. 2022, 11, e220322. [Google Scholar] [CrossRef]
  134. EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA); Turck, D.; Castenmiller, J.; de Henauw, S.; Hirsch-Ernst, K.I.; Kearney, J.; Knutsen, H.K.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Scientific opinion on the relationship between intake of alpha-lipoic acid (thioctic acid) and the risk of insulin autoimmune syndrome. EFSA J. 2021, 19, e06577. [Google Scholar] [CrossRef]
  135. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef] [PubMed]
  136. Gruszczyńska, M.; Sadowska-Przytocka, A.; Szybiak, W.; Więckowska, B.; Lacka, K. Insulin Resistance in Patients with Acne Vulgaris. Biomedicines 2023, 11, 2294. [Google Scholar] [CrossRef]
  137. Monte-Serrano, J.; Villagrasa-Boli, P.; Cruañes-Monferrer, J.; Arbués-Espinosa, P.; Martínez-Cisneros, S.; García-Gil, M.F. Metformina en el tratamiento de enfermedades dermatológicas: Una revisión narrativa [The role of metformin in the treatment of dermatological diseases: A narrative review]. Aten. Primaria 2022, 54, 102354. [Google Scholar] [CrossRef]
  138. Sadati, M.S.; Yazdanpanah, N.; Shahriarirad, R.; Javaheri, R.; Parvizi, M.M. Efficacy of metformin vs. doxycycline in treating acne vulgaris: An assessor-blinded, add-on, randomized, controlled clinical trial. J. Cosmet. Dermatol. 2023, 22, 2816–2823. [Google Scholar] [CrossRef]
  139. Andreadi, A.; Muscoli, S.; Tajmir, R.; Meloni, M.; Minasi, A.; Muscoli, C.; Ilari, S.; Mollace, V.; Della Morte, D.; Bellia, A.; et al. Insulin Resistance and Acne: The Role of Metformin as Alternative Therapy in Men. Pharmaceuticals 2022, 16, 27. [Google Scholar] [CrossRef]
  140. Tam, H.T.X.; Thuy, L.N.D.; Vinh, N.M.; Anh, T.N.; Van, B.T. The Combined Use of Metformin and Methotrexate in Psoriasis Patients with Metabolic Syndrome. Dermatol. Res. Pract. 2022, 2022, 9838867. [Google Scholar] [CrossRef]
  141. Malavazos, A.E.; Meregalli, C.; Sorrentino, F.; Vignati, A.; Dubini, C.; Scravaglieri, V.; Basilico, S.; Boniardi, F.; Spagnolo, P.; Malagoli, P.; et al. Semaglutide therapy decreases epicardial fat inflammation and improves psoriasis severity in patients affected by abdominal obesity and type-2 diabetes. Endocrinol. Diabetes Metab. Case Rep. 2023, 2023, 23-0017. [Google Scholar] [CrossRef]
  142. Yen, H.; Chang, Y.T.; Yee, F.J.; Huang, Y.C. Metformin Therapy for Acne in Patients with Polycystic Ovary Syndrome: A Systematic Review and Meta-analysis. Am. J. Clin. Dermatol. 2021, 22, 11–23. [Google Scholar] [CrossRef]
  143. Kim, J.; Ahn, C.W.; Fang, S.; Lee, H.S.; Park, J.S. Association between metformin dose and vitamin B12 deficiency in patients with type 2 diabetes. Medicine 2019, 98, e17918. [Google Scholar] [CrossRef]
  144. Zacchè, M.M.; Caputo, L.; Filippis, S.; Zacchè, G.; Dindelli, M.; Ferrari, A. Efficacy of myo-inositol in the treatment of cutaneous disorders in young women with polycystic ovary syndrome. Gynecol. Endocrinol. 2009, 25, 508–513. [Google Scholar] [CrossRef] [PubMed]
  145. Radu, A.; Tit, D.M.; Endres, L.M.; Radu, A.F.; Vesa, C.M.; Bungau, S.G. Naturally derived bioactive compounds as precision modulators of immune and inflammatory mechanisms in psoriatic conditions. Inflammopharmacology 2025, 33, 527–549. [Google Scholar] [CrossRef] [PubMed]
  146. Yin, J.; Xing, H.; Ye, J. Efficacy of berberine in patients with type 2 diabetes mellitus. Metabolism 2008, 57, 712–717. [Google Scholar] [CrossRef] [PubMed]
  147. Rao, P.V.; Gan, S.H. Cinnamon: A multifaceted medicinal plant. Evid Based Complement Alternat Med. 2014, 2014, 642942. [Google Scholar] [CrossRef]
  148. Wickenberg, J.; Ingemansson, S.L.; Hlebowicz, J. Effects of green tea and its polyphenols on insulin sensitivity. Eur. J. Clin. Nutr. 2010, 64, 1243–1249. [Google Scholar] [CrossRef]
  149. Zhang, Q.; Xiao, X.; Li, M. Berberine moderates glucose and lipid metabolism through multi-target regulation. Evid.-Based Complement. Altern. Med. 2011, 2011, 924851. [Google Scholar] [CrossRef]
  150. Sheng, X.; Zhang, Y.; Gong, Z.; Huang, C.; Zang, Y.Q. Improved Insulin Resistance and Lipid Metabolism by Cinnamon Extract through Activation of Peroxisome Proliferator-Activated Receptors. PPAR Res. 2008, 2008, 581348. [Google Scholar] [CrossRef]
  151. Posadzki, P.; Watson, L.; Ernst, E. Herb-drug interactions: An overview of systematic reviews. Br. J. Clin. Pharmacol. 2013, 75, 603–618. [Google Scholar] [CrossRef]
  152. Colletti, A.; Fratter, A.; Pellizzato, M.; Cravotto, G. Nutraceutical Approaches to Dyslipidaemia: The Main Formulative Issues Preventing Efficacy. Nutrients 2022, 14, 4769. [Google Scholar] [CrossRef]
Figure 1. Pathogenic crosstalk between systemic insulin resistance (IR) and skin homeostasis disruption in inflammatory dermatoses. Systemic IR leads to hyperinsulinemia and elevated insulin-like growth factor-1 (IGF-1), which converge on key intracellular pathways such as mammalian target of rapamycin complex (mTORC)1 and p38 mitogen-activated protein kinases (MAPK) within the skin. Hyperactivation of mammalian target of mTORC1 fosters basal keratinocyte proliferation while repressing FoxO1, a critical transcription factor for keratinocyte differentiation and lipid synthesis, thereby impairing epidermal barrier formation. Concurrently, p38MAPK activation, fueled by pro-inflammatory cytokines like tumor necrosis factor (TNF)-α and interleukin (IL)-6, amplifies keratinocyte hyperproliferation and suppresses terminal differentiation, exacerbating cutaneous inflammation and disrupting skin integrity. These dysregulated signaling cascades are further compounded by oxidative stress and lipid dysmetabolism, creating a self-sustaining loop that underpins the pathogenesis of IR-driven dermatoses such as psoriasis, acne vulgaris, hidradenitis suppurativa. ↑ = increase; ↓ = reduction.
Figure 1. Pathogenic crosstalk between systemic insulin resistance (IR) and skin homeostasis disruption in inflammatory dermatoses. Systemic IR leads to hyperinsulinemia and elevated insulin-like growth factor-1 (IGF-1), which converge on key intracellular pathways such as mammalian target of rapamycin complex (mTORC)1 and p38 mitogen-activated protein kinases (MAPK) within the skin. Hyperactivation of mammalian target of mTORC1 fosters basal keratinocyte proliferation while repressing FoxO1, a critical transcription factor for keratinocyte differentiation and lipid synthesis, thereby impairing epidermal barrier formation. Concurrently, p38MAPK activation, fueled by pro-inflammatory cytokines like tumor necrosis factor (TNF)-α and interleukin (IL)-6, amplifies keratinocyte hyperproliferation and suppresses terminal differentiation, exacerbating cutaneous inflammation and disrupting skin integrity. These dysregulated signaling cascades are further compounded by oxidative stress and lipid dysmetabolism, creating a self-sustaining loop that underpins the pathogenesis of IR-driven dermatoses such as psoriasis, acne vulgaris, hidradenitis suppurativa. ↑ = increase; ↓ = reduction.
Ijms 26 07538 g001
Figure 2. Canonical insulin signaling pathways and their impairment in insulin resistance. Upon insulin binding, the insulin receptor (IR) undergoes autophosphorylation and activates insulin receptor substrates (IRS-1/2), leading to activation of PI3K and conversion of PIP2 to PIP3. This recruits Akt and PDK1 to the membrane, where Akt is phosphorylated and fully activated by PDK1 and mTORC2. Activated Akt promotes glucose uptake via GLUT-4 translocation, glycogen synthesis, and nitric oxide (NO) production through eNOS activation. Parallel activation of the MAPK pathway regulates transcriptional programs for cell proliferation. In IR, inflammatory cytokines (e.g., TNF-α, IL-6), free fatty acids (FFAs), and reactive oxygen species (ROS) induce serine phosphorylation of IRS-1/2, impairing PI3K activation and GLUT-4 translocation, ultimately reducing glucose uptake and contributing to IR. ↑ = increase; ↓ = reduction; red dashed line = inhibition.
Figure 2. Canonical insulin signaling pathways and their impairment in insulin resistance. Upon insulin binding, the insulin receptor (IR) undergoes autophosphorylation and activates insulin receptor substrates (IRS-1/2), leading to activation of PI3K and conversion of PIP2 to PIP3. This recruits Akt and PDK1 to the membrane, where Akt is phosphorylated and fully activated by PDK1 and mTORC2. Activated Akt promotes glucose uptake via GLUT-4 translocation, glycogen synthesis, and nitric oxide (NO) production through eNOS activation. Parallel activation of the MAPK pathway regulates transcriptional programs for cell proliferation. In IR, inflammatory cytokines (e.g., TNF-α, IL-6), free fatty acids (FFAs), and reactive oxygen species (ROS) induce serine phosphorylation of IRS-1/2, impairing PI3K activation and GLUT-4 translocation, ultimately reducing glucose uptake and contributing to IR. ↑ = increase; ↓ = reduction; red dashed line = inhibition.
Ijms 26 07538 g002
Figure 3. Molecular mechanisms of insulin sensitizing agents (ISAs) targeting insulin resistance, oxidative stress, and inflammation in skin disorders. The figure illustrates how nutraceutical ISAs, including myo-inositol, alpha-lipoic acid, vitamin D, vitamin C, folic acid, and B vitamins, modulate insulin signaling pathways and inflammatory responses implicated in cutaneous insulin resistance. Myo-inositol and alpha-lipoic acid enhance insulin signaling through IRS-1 and Akt, improving glucose metabolism and reducing oxidative stress via AMPK activation. Vitamin D and alpha-lipoic acid suppress NF-κB-mediated inflammation, while vitamin C supports collagen synthesis and combats reactive oxygen species (ROS). Folic acid and B vitamins contribute to endothelial function and homocysteine metabolism, indirectly alleviating systemic and cutaneous inflammation. These coordinated actions mitigate keratinocyte dysfunction, oxidative stress, and chronic inflammation, which are central to the pathogenesis of insulin resistance-associated skin diseases such as psoriasis, acne, hidradenitis suppurativa, and acanthosis nigricans.
Figure 3. Molecular mechanisms of insulin sensitizing agents (ISAs) targeting insulin resistance, oxidative stress, and inflammation in skin disorders. The figure illustrates how nutraceutical ISAs, including myo-inositol, alpha-lipoic acid, vitamin D, vitamin C, folic acid, and B vitamins, modulate insulin signaling pathways and inflammatory responses implicated in cutaneous insulin resistance. Myo-inositol and alpha-lipoic acid enhance insulin signaling through IRS-1 and Akt, improving glucose metabolism and reducing oxidative stress via AMPK activation. Vitamin D and alpha-lipoic acid suppress NF-κB-mediated inflammation, while vitamin C supports collagen synthesis and combats reactive oxygen species (ROS). Folic acid and B vitamins contribute to endothelial function and homocysteine metabolism, indirectly alleviating systemic and cutaneous inflammation. These coordinated actions mitigate keratinocyte dysfunction, oxidative stress, and chronic inflammation, which are central to the pathogenesis of insulin resistance-associated skin diseases such as psoriasis, acne, hidradenitis suppurativa, and acanthosis nigricans.
Ijms 26 07538 g003
Figure 4. Intestinal absorption of myo-inositol, role of magnesium as positive effector of inositol transport, and myo-inositol effects on insulin resistance.
Figure 4. Intestinal absorption of myo-inositol, role of magnesium as positive effector of inositol transport, and myo-inositol effects on insulin resistance.
Ijms 26 07538 g004
Table 1. Clinical studies conducted with oral MI in dermatological diseases.
Table 1. Clinical studies conducted with oral MI in dermatological diseases.
Author(s), YearStudy DesignParticipantsInterventionPrimary OutcomeAdverse EventsFollow-Up (Weeks)
Pezza et al., 2015 [121]RCT50 women with PCOS and acneLEVIGON MI 2 g, microlipodispersed magnesium 56.25 mg, folic acid 200 mcg BIDDecreased number of papulopustular lesionsNone6
Pezza et al., 2025 [122]Retrospective study200 women with PCOS and acneLEVIGON MI 2 g, microlipodispersed magnesium 56.25 mg, folic acid 200 mcg BIDReduction in BMI, testosterone, free testosterone, and DHEAS levels, Improvement of quality of life (Cardiff Acne Disability Index and Dermatology Life Quality Index) and Ferriman–Gallwey scoreNone24
Fruzzetti et al., 2017 [123]RCT50 women with PCOSMI 4 g, folic acid 400 mcgReduction in BMI, 20% felt improvement in hirsutism; 38% in acneNone6
Minozzi et al., 2008 [124]Uncontrolled clinical trial46 women with mild to moderate hirsutismMI 2 g, BIDHirsutism score decreased by −2.3 ± 0.9 (p < 0.001)None6
Advani et al., 2019 [125]Retrospective trial51 women (35 obese, 16 lean) with PCOSTrazer F Forte, BID (inositol MI:DCI 600 mg, NAC 300 mg, Biotin 5 mg, Lycopene 5 mg, vitamin D 400IU)Acne scores decreased significantly in obese patients and in lean patients)None3
Ramanan et al., 2020 [126]Uncontrolled clinical trial32 women with mild to moderate acne and hirsutismTracnil, BID (MI 2 g, folic acid 1 mg, vit D3 1000 IU)Significant improvement in GA and hirsutism scores (p < 0.05)Mild GI distress in some patients6
Allan et al., 2004 [127]Crossover RCT23 patients with psoriasisInositol 6 g, QDPatients on lithium: PASI scores improved (p > 0.05); patients not on lithium: PASI scores improved significantly (p = 0.015)None2.5
Donnarumma et al., 2020 [128]RCT10 patients with HSAntibiotics + MI 2 g, liposomal magnesium, folic acid, BIDReduction in Sartorius scores (p < 0.04)None6
Abbreviations: PCOS: polycystic ovarian syndrome; MI: myo-inositol; DCI: d-chiro-inositol; QD: once daily; BID: twice daily; PASI: psoriasis area and severity index; HS: hidradenitis suppurativa.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Morrone, P.; Caroppo, F.; De Pedrini, A.; Colletti, A.; Baj, G. New Perspectives on Nutraceutical Insulin Sensitizing Agents in the Treatment of Psoriasis and Other Dermatological Diseases. Int. J. Mol. Sci. 2025, 26, 7538. https://doi.org/10.3390/ijms26157538

AMA Style

Morrone P, Caroppo F, De Pedrini A, Colletti A, Baj G. New Perspectives on Nutraceutical Insulin Sensitizing Agents in the Treatment of Psoriasis and Other Dermatological Diseases. International Journal of Molecular Sciences. 2025; 26(15):7538. https://doi.org/10.3390/ijms26157538

Chicago/Turabian Style

Morrone, Pietro, Francesca Caroppo, Alberto De Pedrini, Alessandro Colletti, and Germano Baj. 2025. "New Perspectives on Nutraceutical Insulin Sensitizing Agents in the Treatment of Psoriasis and Other Dermatological Diseases" International Journal of Molecular Sciences 26, no. 15: 7538. https://doi.org/10.3390/ijms26157538

APA Style

Morrone, P., Caroppo, F., De Pedrini, A., Colletti, A., & Baj, G. (2025). New Perspectives on Nutraceutical Insulin Sensitizing Agents in the Treatment of Psoriasis and Other Dermatological Diseases. International Journal of Molecular Sciences, 26(15), 7538. https://doi.org/10.3390/ijms26157538

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