Abstract
Hair loss is a global concern, driving substantial public interest in effective therapeutic solutions. Beyond its protective physiological functions, hair plays a pivotal role in social and non-verbal communication, which can be profoundly disrupted by hair loss. While conventional pharmacological treatments are commonly employed, their use is often limited by the risk of adverse effects. To address this limitation, we propose the exploration of phytochemicals derived from natural sources as safer and potentially effective alternatives for hair loss prevention. Many natural compounds have shown the capacity to activate key signaling pathways involved in hair growth, offering promising avenues for innovation in the hair industry. This study explores the biological mechanisms underlying hair development, morphogenesis, and regeneration, with a particular focus on the Wnt/β-catenin, Sonic hedgehog (Shh), and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathways, which are critical to hair growth. Furthermore, we provide a comprehensive compilation of natural materials known to promote hair growth and their associated phytochemicals. This repository serves as a foundation for identifying and developing novel agents to enhance hair regeneration. Our review highlights the need for continued research into identifying and refining safe, naturally derived candidates for the prevention and treatment of hair loss, thereby addressing a pressing unmet medical and cosmetic need.
Keywords: Hair, hair follicle, pathways, phytochemicals, hair growth
Introduction
Mammals exhibit traits that differentiate them from non-mammals, with hair being one of the most distinctive. In humans, hair growth occurs on all skin surfaces except palmoplantar regions, such as the palms and soles [1], and hair plays an essential role in homeostasis by providing protection against external damage and contributing to thermoregulation. For example, scalp hair shields the head and neck, while eyelashes and eyebrows prevent contaminants from entering the eyes [2]. Hair also serves sensory functions, enhancing the skin’s ability to detect sensory stimuli [3]. Moreover, hair serves as a medium for social communication, and accordingly, disorders affecting hair can lead to considerable psychological distress, manifesting as depression, diminished confidence, and anxiety. Notably, personality disorders are more prevalent among men experiencing hair loss than among women experiencing hair loss [4].
The condition of hair is influenced by multiple factors, including sex steroid hormones, stress, ultraviolet (UV) exposure, smoking, microbial infections, environmental pollutants, irritants, and chemical exposure. Among these, sex hormones, particularly androgens, play a central role in androgenetic alopecia (AA), a common form of hair loss [5]. Additionally, exogenous factors that induce oxidative stress have deleterious effects on hair growth and retention [6]. Consequently, mitigating these detrimental factors is vital for maintaining hair health.
Currently, a range of hair growth treatments, including oral medications, topical applications, platelet-rich plasma therapy, laser therapy, and microneedling, is available [7]. Conventional hair growth therapies frequently target the regulation of sex hormones, but these are limited by significant side effects [8]. This underscores an urgent need for the development of safe and effective alternatives that minimize side effects. Botanical products have emerged as promising alternatives to synthetic treatments for preventing hair loss and promoting hair growth. Many plants contain diverse phytochemicals that synergistically enhance hair health [9]. Importantly, phytochemicals derived from botanical sources generally exhibit fewer and less acute toxic effects than synthetic drugs.
This review focuses on phytochemicals targeting hair growth-related molecular pathways as an innovative strategy for promoting hair health. We outline hair development processes, identify candidate compounds that modulate specific signaling pathways, and suggest ways to minimize side effects. The natural product-derived compounds regulating these cascades offer a promising and safe approach to effective hair growth treatments.
General Description of Hair Development
The Role of Hair
Hair originates at the base of the hair follicle (HF), which serves as the anchoring structure connecting the hair to the skin. Hair performs physiological functions, helping to retain body heat, facilitate sweating, and disperse sebaceous secretions, as well as psychological functions [10]. Initial hairs, termed “lanugo,” develop during the fetal period. During childhood, hair is classified as “vellus,” whereas by adulthood, it has transitioned to “terminal” hair. Each stage exhibits distinct characteristics in terms of color, length, diameter, and hardness [11]. Hair growth occurs at an average rate of 10–15 mm per month. From childhood onwards, humans possess approximately 5 million hairs, with roughly 100,000 HFs located on the scalp [12].
Composition and Morphology of the HF
Hair follicles, highly conserved skin appendages, function as complex mini-organs with critical physiological roles primarily related to hair growth. Located within the dermis, HFs are among the few organs in the human body capable of regeneration throughout life [10, 13]. The terminal HF has a total length of 3,864 ± 605 μm and a diameter of 172 ± 70 μm [14].
The histomorphology of the HF is illustrated in Fig. 1, adapted from Schneider et al. [15]. Mature HFs are anatomically divided into two main regions: the upper and lower regions. The upper region comprises the infundibulum and isthmus, while the lower region includes the suprabulbar area and the bulb. The bulge, located within the permanent region, is a non-cycling zone. The permanent region also encompasses the arrector pili muscle and sebaceous glands, whereas the cycling region contains the dermal papilla (DP) and matrix. The DP, derived from mesodermal origin, influences the diameter and length of the hair shaft (HS), the size of the hair bulb (HB), and the duration of the growth phase [16, 17]. A longitudinal section of the HF reveals a cylindrical structure composed of eight distinct layers [18]. From the outermost to the innermost layer, the HF is organized into four major components: the inner root sheath (IRS), the outer root sheath (ORS), the associated layer, and the HS. The IRS is further subdivided into Henle’s layer, Huxley’s layer, and the IRS cuticle [19]. The HS itself consists of the medulla, cortex, and stratum corneum [20].
Fig. 1. Histomorphology of the hair follicle.
HF Morphogenesis and Regeneration
Hair follicle morphogenesis and regeneration are typically considered as two interrelated phenomena [21]. Hair follicle morphogenesis predominantly occurs during embryonic development and involves intricate interactions between mesenchymal and epithelial tissues [22, 23]. While this process is primarily confined to the fetal stage, it has been demonstrated that new HFs can be induced postnatally through the transplantation of human HF dermal sheath tissue [24]. The progression of HF formation is delineated into eight specific stages, as outlined in Section HF Morphogenesis.
Upon the conclusion of HF morphogenesis, the follicle transitions into the regeneration phase, beginning with the formation of the HS. This phase is initiated by the shedding of the primary HS, which is subsequently replaced by a newly formed HS. Hair follicle regeneration commences during the catagen phase, which is marked by the postpartum shedding of lanugo hair and the emergence of vellus hair [25].
The prevailing consensus within the field is that HF regeneration proceeds through three cyclic stages: catagen (regression), telogen (quiescence), and anagen (growth) [11]. However, some researchers have proposed an alternative model incorporating a four-stage cycle, which includes the exogen phase, in which the existing mature HS is shed, as an independent and active process distinct from the telogen phase [26-28]. A key point of debate is whether the exogen phase is a passive component of telogen or represents a distinct, physiologically regulated stage. Milner et al. [28] provided evidence for the latter, demonstrating that hair shedding in mice involves active physiological regulation via protein hydrolysis. Further refinement of this concept was provided by Higgins et al.[23], who subdivided the exogen phase into “early exogen” and “late exogen.” Their analysis of mouse vibrissa follicles from the early anagen to late exogen stages revealed a progressive reduction in tissue attached to the club fiber, with minimal or no cellular material present in late exogen. These findings suggest that proteolysis facilitates the separation of the club fiber from the ORS, thereby enabling fiber shedding [29]. Moreover, experiments using desmoglein 3 gene-disrupted mice demonstrated that the absence of this gene leads to significant hair loss during telogen, underscoring its crucial role in anchoring hairs to the HF ORS [30]. Collectively, these findings support the notion that protein hydrolysis drives a transition from the telogen to a distinct exogen stage, reinforcing the concept of exogen as an autonomous and active stage of HF regeneration. Nevertheless, this interpretation remains a subject of ongoing debate.
Over the course of an individual’s lifetime, HFs undergo approximately 10–20 complete cycles, with scalp follicles cycling asynchronously [31]. Importantly, the hair growth cycle can be influenced by various exogenous factors, including hair plucking, skin trauma, temperature fluctuations, laser stimulation, and hormonal changes, which can accelerate or delay HF growth and influence hair vitality [32, 33].
HF Morphogenesis
Hair follicle morphogenesis in mammals typically occurs once during the embryonic stage [34]. However, emerging evidence indicates that de novo HF formation can occur following wounding, as reported in several studies [35, 36]. Hair follicle formation is a dynamic process initiated by intricate signaling interactions between dermal mesenchymal and epithelial cells during embryogenesis [37]. It is widely accepted that the dermal mesenchyme functions as the inducer of HF formation, while the epithelium acts as the responder [38]. At approximately 50–60 days of gestation, epithelial cells respond to dermal signaling cues by forming the initial HF structure, referred to as the placode. These placode cells then transmit reciprocal signals to the underlying dermal mesenchyme, inducing dermal agglutination, a process primarily mediated by Wnt/β-catenin signaling pathways [37, 39].
Next, responding to Sonic hedgehog (Shh) signals from the dermal agglutination cluster, the epidermal placode invaginates into the dermis to form the primary hair germ. As growth progresses, the hair peg develops, culminating in the formation of the bulb, as aggregated dermal cells differentiate into the DP [39]. Signaling from the DP then stimulates rapid epithelial cell division, resulting in the encapsulation of the DP by HF epithelial cells and the formation of the HB. As the HF extends downward, the hair matrix (HM) undergoes rapid proliferation, migrates upward, and differentiates into the IRS and HS [40]. Concurrently, accessory HF structures, including the sebaceous glands and arrector pili muscles, are fully developed. At this stage, the differentiated HS begins to emerge from the scalp, with fully mature HFs producing the fine lanugo hair observed at birth [41].
Anatomical studies of rodent and human HFs have categorized HF development into eight distinct stages. Table 1 and Fig. 2, synthesized from multiple published studies and reviews, delineate the defining characteristics of each stage of HF morphogenesis [15, 25, 42-48].
Table 1.
Key features of hair follicle morphogenesis.
|
Fig. 2. Overview of the hair follicle morphogenesis process.
HF Regeneration
Hair follicles are cycling structures, undergoing continuous regenerative cycles throughout adult life, that serve as both stem cell reservoirs and HS factories [49-51]. While HF morphogenesis (embryonic organogenesis) and regeneration (postnatal regrowth) share several characteristics, they are distinct processes [21]. During regeneration, most HF structures arise from the inner matrix of the HB, with the ORS, derived from bulge cells, being the only exception [52]. In anagen, the ORS migrates downward as the IRS and HS ascend [53].
Hair follicle regeneration is conventionally divided into three or four phases (Section 1.3), which can be further subdivided into multiple sub-phases. Using a murine model, Müller-Röver et al. [54] meticulously classified HFs at various stages, segmenting the anagen and catagen phases into eight sub-stages based on HF length and skin thickness. The HF’s length is shortest during telogen, increases to a maximum in anagen VI, remains stable until catagen II, and then shortens until the next telogen phase starts (Fig. 2) [54]. Each stage exhibits distinct characteristics, enabling accurate identification [25, 54, 55]. Table 2 summarizes these features, and Fig. 3 depicts them visually.
Table 2.
Key features of hair follicle regeneration.
|
Fig. 3. Overview of the hair follicle regeneration process.
Catagen: The Regression Phase
The catagen phase, the initial stage following HF morphogenesis, typically lasts approximately two weeks [47]. A defining feature of this phase is the formation of the club hair (CH), marking the separation of the HB from the DP [56]. Additional hallmarks of this phase include the cessation of terminal differentiation, apoptosis of keratinocytes, and halting of protein and melanin production within the regressing HB [57]. These prominent morphological changes distinctly signal the onset of catagen.
Telogen: The Resting Phase
The identification of HFs in the telogen phase is relatively straightforward. In this stage, HFs are fully atrophied and located entirely within the dermis, with new HS formation not yet initiated [47]. Historically, telogen has been regarded as a resting phase; however, emerging evidence suggests ongoing physiological activity during this period. For example, expression levels of Cadherin 13 [58], Desmoglein 3 [30], and Keratin 24 [59] reach their peak during telogen.
Exogen: The Active Shedding Phase
Traditionally, the mature HS is considered to be shed at the conclusion of the telogen phase or during the onset of the anagen phase (sub-stages anagen I–IV). However, as detailed above, some researchers classify this shedding process as a distinct phase termed “exogen,” aligning with the nomenclatural convention of the other phases [28, 51]. Recent studies have confirmed the presence of the exogen stage in the human scalp hair cycle. Unlike the telogen phase, in which hairs remain firmly anchored to the scalp, as they do in the anagen phase, during the exogen phase, hairs are easily removed from the HF without any attached cellular structures [23, 60].
Anagen: The Growth Phase
The transition from telogen to anagen is marked by significant morphological and cellular changes. The pre-anagen phase closely resembles the late telogen, with a notable distinction being the enlargement and downward growth of the DP. Simultaneously, stem cells within the HF bulge rapidly proliferate, differentiating into new HS cells [25].
Throughout the anagen phase, HF keratinocytes exhibit vigorous proliferation, progressing through multiple sub-stages that culminate in the formation of the ORS, IRS, and HS. This phase typically spans 3–5 years, with hair growth occurring at an average rate of approximately 0.3 mm per day [61].
The regenerative capacity of HFs has been explored through several theoretical frameworks, including the epithelial theory, oscillating signal theory, bulge activation theory, papilla morphogen theory, resonance theory, inherent embryonic cycle theory, and inhibition-disinhibition theory [62]. However, the precise regulatory mechanisms underlying HF regeneration remain to be fully elucidated.
Regulation of Hair Regeneration
Hair morphogenesis, generation, and growth are orchestrated by a complex interplay of multiple signaling pathways, including Hedgehog, Wnt, and bone morphogenetic proteins (Bmp2, Bmp4), which are essential for HF induction and morphogenesis [63]. These pathways interact synergistically to sustain the hair cycle, whereas their aberrant activation can cause hair dysplasia and skin cancer [64]. Despite considerable advances in this field, the precise mechanisms underlying HF formation and HF stem cell (HFSC) regulation remain incompletely understood.
Wnt/β-catenin Signaling
The Wnt/β-catenin signaling pathway is initiated by the binding of Wnt proteins to cell membrane receptors [65]. In the absence of Wnt ligands, β-catenin is phosphorylated by glycogen synthase kinase-3β (GSK3β), axis inhibition protein (Axin), adenomatous polyposis coli (APC), and casein kinase 1 (CK1) within the destruction complex, leading to proteasomal degradation [66]. Afterwards, the Groucho/transducin-like enhancer of split (TLE) suppresses the lymphoid enhancer factor (LEF)/T cell factor (TCF)-mediated transcription of hair growth-related genes [67].
Testosterone exerts a critical inhibitory effect on the Wnt/β-catenin signaling pathway. Upon entry into the cell, testosterone is converted to dihydrotestosterone (DHT) by 5α-reductase (5AR). Then, DHT binds to the androgen receptor (AR) and induces dickkopf-related protein 1 (DKK1) expression [68], suppressing HFSC differentiation and triggering catagen [69] (Fig. 4A).
Fig. 4. Key signaling pathways involved in the regulation of hair regeneration:
the (A) inactivated and (B) activated states of the Wnt/β-catenin signaling pathway, (C) activated and inactivated states of the Shh signaling pathway, and (D) activated and inactivated states of the JAK-STAT signaling pathway.
Conversely, Wnt activation promotes the disheveled (DVL)-mediated inhibition of the dissemination of the destruction complex, enabling β-catenin to translocate to the nucleus and bind to transducin β-like protein 1 (TBL1) and TBL1-related protein (TBLR1). This complex, in turn, binds to LEF/TCF transcription factors, displacing Groucho/TLE1 and histone deacetylase 1 (HDAC1) to initiate target gene transcription [67, 70, 71]. Additionally, 5AR inhibition also prevents DHT-mediated DKK1 expression, sustaining hair growth [69] (Fig. 4B).
Sonic Hedgehog Signaling in Hair Regeneration
The Shh signaling pathway is pivotal in HF development, with Shh being expressed in presumptive HFs [72]. In the absence of the Shh ligand, the 12-pass transmembrane receptor patched (PTCH) inhibits smoothened (SMO) activity [73]. When present, the Shh ligand binds to PTCH, relieving this inhibition and allowing the activation of SMO and the downstream glioma-associated oncogene homolog (GLI) family of transcription factors regulated by the suppressor of fused (SUFU) [74, 75]. By promoting the proteolytic cleavage of GLI2/3 into their repressor forms (GLI2/3R) that inhibit target gene transcription, SUFU functions as a negative regulator [76, 77]. Upon activation, GLI proteins translocate to the nucleus to induce the expression of genes (PTCH1, GLI1, cyclin D1, cyclin D2, and Sox9), crucial for HFSC maintenance [78, 79]. Experimental studies have shown that wild-type nude mice expressing the Shh gene continue to develop hair, whereas Shh mutants remain hairlessness and with pigmented skin [80]. The comprehensive Shh pathway is depicted in detail in Fig. 4C.
JAK-STAT Signaling in Hair Regeneration
The JAK-STAT signaling pathway is implicated in numerous pathological conditions, including malignancies and autoimmune diseases, such as AA [81-84]. JAK kinases -JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2)- phosphorylate STAT transcription factors upon cytokine- or growth factor-induced receptor dimerization [81, 85]. Phosphorylated STATs dissociate from the receptor, form homo- or heterodimers, and translocate to the nucleus [86]. In the nucleus, STAT dimers bind to specific DNA sequences to regulate gene expression, acting as transcriptional complexes. These transcriptional events ultimately inhibit hair growth [87], contributing to the pathogenesis of AA [84] (Fig. 4D). Thus, the modulation of JAK-STAT signaling represents a promising therapeutic strategy for addressing hair growth disorders.
Mechanisms of Action and Adverse Effects of Hair Loss Management Drugs
Current pharmacological treatments for AA either promote hair growth or prevent hair loss, but their adverse effects necessitate a thorough understanding of their mechanisms of action and potential risks [88]. This section categorizes these drugs, summarizes their therapeutic efficacy, and outlines associated risks.
Vasodilators
Vasodilators relax vascular smooth muscle, enhancing blood flow to regions with oxygen and nutrient deficiencies and supporting angiogenesis [89, 90]. Given the association between vasodilation and angiogenesis [91], promoting vascular dilation holds promise for stimulating hair growth. Topically administered minoxidil, a vasodilator-based treatment, has mechanisms of action that remain only partially elucidated. Minoxidil enhances the β-catenin signaling pathway in human dermal papilla cells (hDPCs), prolongs the anagen phase, and mitigates the transition to catagen in murine models, thereby promoting anagen phase maintenance [92]. It also downregulates AR-related functions [93], influences cell proliferation and apoptosis in hDPCs [94], and promotes root sheath outgrowth along the hair shaft in murine vibrissae follicle culture models [95]. Despite its well-documented efficacy, minoxidil is associated with adverse effects, including local erythema and pruritus, as well as other clinical symptoms [96-98].
5AR Inhibitors
As their name suggests, 5AR inhibitors specifically target 5AR, the enzyme responsible for the conversion of testosterone to DHT, a key androgen implicated in the pathogenesis of AA [99]. FDA-approved 5AR inhibitors effectively reduce the effects of DHT by inhibiting 5AR enzymatic activity [100].
Finasteride is an antiandrogenic agent that targets type II 5AR, facilitating hair regeneration by upregulating Wnt/β-catenin signaling through AKT phosphorylation and β-catenin activation. Additionally, finasteride enhances the expression of self-renewal transcription factors, such as Sox-2 and Nanog, in DP cells, promoting their aggregation and a stem cell-like phenotype [101, 102]. In animal studies, finasteride significantly reduced serum DHT levels and increased the number of anagen-phase follicles, which resulted in longer follicles and increased hair weights [103]. However, its use has been linked to severe adverse effects, including sexual dysfunction, orthostatic hypotension, and dizziness [104].
Unlike finasteride, dutasteride, another 5AR inhibitor, inhibits both type I and type II isoenzymes, resulting in a more pronounced suppression of serum DHT levels, as demonstrated in clinical studies [101]. Dutasteride effectively inhibited the formation of 5α-DHT, a testosterone metabolite, thereby mitigating AA [105]. Furthermore, dutasteride increases hair length and the proportion of HSs and HFs in the anagen phase [106]. Despite its therapeutic efficacy, dutasteride may cause sexual dysfunction (e.g., decreased libido and impotence) and gynecomastia [88, 104].
AR Antagonists
Dihydrotestosterone demonstrates a greater binding affinity for the ARs than testosterone, highlighting the critical role of ARs in androgen signaling pathways [107], and AR antagonists prevent androgens–AR interactions [108]. Additionally, nonsteroidal AR antagonists act through the competitive inhibition of AR binding, further impeding androgen signaling and mitigating hair loss [109].
The AR antagonist spironolactone reduces testosterone levels and blocks AR activity [110]. However, its clinical use is associated with gynecomastia, hyperkalemia, irregular menstruation, muscle cramps, hypotension, and decreased libido [111]. Another AR antagonist, cyproterone acetate, acts as a direct inhibitor of DHT–AR binding, while concurrently reducing the secretion of follicle-stimulating hormone and luteinizing hormone, leading to decreased testosterone levels [110]. It also significantly influences the proliferation of DP cells [112].
Nonsteroidal AR Antagonist
Flutamide is a nonsteroidal AR antagonist that competitively inhibits the binding of DHT to ARs [113, 114]. Preclinical studies in murine models have demonstrated that flutamide treatment promotes hair regrowth and increases HS length and diameter [113, 115]. However, flutamide therapy is associated with libido reduction, gynecomastia, anxiety, rash, and somnolence [116].
PGF2 Analogs
Prostaglandins (PGs) represent promising therapeutic targets for hair loss, with PGE2 and PGF2α acting as potent hair growth stimulators in hair follicles [117]. Initially, PGF2 analogues were utilized for reducing intraocular pressure but were associated with hypertrichosis [118]. This phenomenon has since been leveraged to stimulate hair growth, first demonstrated by the application of PGF2 analogues to murine dorsal skin during both the telogen and anagen phases [119].
Latanoprost, originally approved as an ophthalmic solution [120], has been demonstrated to stimulate keratinocyte proliferation within HFs, thereby facilitating hair growth. Moreover, latanoprost enhances dermal vasodilation, improving HF nutrition through increased blood flow [121]. However, its use has been associated with alterations in eyelash morphology and the development of iridial and periocular hyperpigmentation [122]. A second, bimatoprost effectively increased both the number and length of HFs in the anagen phase in scalp follicle organ culture models and promoted hair regrowth in vivo [123], without notable adverse effects [124].
Imidazole Antifungal Agents
Imidazole compounds, such as ketoconazole, well-known for their broad-spectrum antifungal activity, have also been shown to facilitate hair growth by inhibiting DHT production and/or binding to ARs [125, 126]. Ketoconazole upregulates OVOL1 expression at the cellular level, suggesting that its effects on hair growth may be mediated through the Wnt/β-catenin signaling cascade [127, 128]. Moreover, it has been shown to promote hair regrowth and increase HF diameter in male murine models [129]. Despite these therapeutic benefits, ketoconazole use is associated with decreased libido, nausea, vomiting, pruritus, and gynecomastia [103].
JAK Inhibitors
JAK inhibitors have gained attention as targeted molecular therapies for AA, and the U.S. FDA has approved tofacitinib, ruxolitinib, and ritlecitinib for human use [130, 131]. Tofacitinib, a pan-JAK inhibitor, targets JAK1, JAK2, JAK3, and TYK2, while ruxolitinib specifically inhibits JAK1 and JAK2. Tofacitinib promotes hair follicle dermal papilla cell (HFDPC) growth, upregulates β-catenin and Gli1 expression, and regulates cell cycle progression [132-134]. On the other hand, ruxolitinib upregulates both the Wnt/β-catenin and JAK-STAT pathways. To date, these agents have not been associated with significant adverse effects [130]. Additionally, orally administered baricitinib, a JAK1/2 inhibitor, demonstrated effectiveness in stimulating hair growth in both patients with AA and murine models [135]. Although generally well-tolerated, baricitinib has been associated with transient adverse effects, including neutropenia and a reduced reticulocyte count, as reported in some studies [136]. Another orally administered JAK inhibitor, CTP-543, which has received a Fast Track designation from the FDA, has shown promise in promoting the anagen phase of HFs by inhibiting JAK1 and JAK2 [137]. Additional emerging agents, including ritlecitinib (PF-06651600) and brepocitinib (PF-06700841), have demonstrated the potential to facilitate hair regrowth through the inhibition of JAK activation [132, 137-139].
As previously discussed, numerous pharmacological agents carry the risk of serious systemic side effects. Consequently, ongoing research is focused on identifying drugs with a more favorable safety profiles, with a particular focus on those that promote hair growth via the Wnt/β-catenin or Shh signaling pathways [140] or by inhibiting the JAK-STAT pathway [132].
Natural Nutraceuticals/Cosmeceuticals Affecting Hair Growth Signaling Pathways
Current pharmacological treatments for hair loss are effective but frequently associated with undesirable side effects, particularly sexual dysfunction. This has driven interest in safer alternatives that can promote hair growth without such complications. Offering a safer and potentially more sustainable approach, natural nutraceuticals and cosmeceuticals are increasingly recognized as promising candidates.
Hair Growth-Promoting Natural Products and Their Mechanisms of Action
This section examines a range of natural extracts with demonstrated hair growth-promoting properties, underscoring their potential as safer alternatives to conventional treatments (Table 3).
Table 3.
Mechanisms of action of extracts promoting hair growth.
|
The propolis derived from Philippines stingless bees has shown significant hair growth-promoting effects in murine HFs, increasing the number of HFs in the anagen phase with greater efficacy than minoxidil. Mechanistically, this propolis activated both the mRNA and protein expressions of key components within the Wnt/β-catenin signaling pathway, including Lef1, Wnt3a, β-catenin, and Bmp2 [141].
Gardenia florida fruit extract was shown to stimulate cell proliferation and upregulate Wnt/β-catenin signaling, vascular endothelial growth factor (VEGF), and TGF-β1 in hDPCs. In murine models, topical application of the extract to the dorsal skin induced hair regeneration and increased hair length [142].
Mangifera indica leaf extract downregulated DKK1 and SRD51A mRNA levels while upregulating the expression of Axin2, NKD1, and Myc. Furthermore, it reduced DKK1 mRNA expression in response to DHT. In vivo, a 1%topical formulation of the extract promoted hair growth and elongation in treated mice [143].
De-saponinated Camellia seed cake extract, a by-product of the Camellia oleifera seed oil extraction process, enhanced DP cell proliferation and facilitated entry into the anagen phase of the hair cycle. Additionally, it activated potassium channels, counteracting the inhibitory effects of tolbutamide (a potassium channel blocker) on DP cell proliferation. At the cellular level, the extract regulated cytokine expression and promoted the phosphorylation of ERK and AKT. Topical applications in a murine model induced hair growth and increased VEGF content in the skin [144].
Centipedegrass extract has been shown to significantly accelerate the hair cycle in hDPCs via activation of the β-catenin pathway. It also promoted cell proliferation, increased ALP expression, and upregulated hair cycle-related genes and β-catenin signaling molecules. In mice, this extract effectively transitioned hair follicles from the telogen to the anagen phase, outperforming minoxidil across all hair growth markers [145].
Malva verticillata extract exhibited cell proliferation activity, and linoleic acid, one of its bioactive components, activated the Wnt/β-catenin pathway, promoting hair regrowth in the presence of DHT [146]. Similarly, Salvia plebeia extract stimulated cell proliferation and upregulated HGF expression in hDPCs [147]. As epithelial cell growth is inhibited by androgen-inducible transforming growth factor-β1 (TGF-β1) [148], suppressing TGF-β1 expression offers a promising approach for promoting sustained hair growth. The S. plebeia extract not only inhibited TGF-β1 expression but also regulated the Wnt/β-catenin pathway and phosphorylated ERK and AKT in hDPCs. Additionally, in C57BL/6 mice, it facilitated hair growth and HF formation [147].
The titrated extract of Centella asiatica has been shown to promote hair growth in 3D spheroid cultures of human HFDPCs by increasing cell proliferation and spheroid diameter. While initial investigations suggested potential activation of Wnt/β-catenin signaling, subsequent findings indicated no effect on genes associated with this pathway. Instead, the extract appears to inhibit STAT activation [149].
The methanolic extract of the Miscanthus sinensis var. purpurascens flower significantly enhanced cell proliferation in hDPCs and activated ERK phosphorylation and β-catenin signaling. Topical application of the extract to telogenic C57BL/6 mice skin resulted in increased black skin pigmentation and extended HF length. Additionally, the extract suppressed the secretion of cytokines implicated in hair growth regulation, including TGF-β1 and VEGF [150].
The topical application of Angelica sinensis induced significant hair regrowth in depilated mice, evidenced by a transition of skin color from pink to grey, the emergence of new hairs, the transition of HFs from the catagen to anagen phase, and the restoration of HF sizes and HS lengths. Additionally, the treatment inhibited cell apoptosis by modulating the NF-κB and MAPK signaling pathways [151].
Crataegus pinnatifida extract was shown to promote hair growth in hDPCs through enhanced cell proliferation and activation of MAPK and AKT phosphorylation. Notably, oral administration of the extract facilitated hair growth and inhibited apoptosis in murine skin tissue more effectively than finasteride [152].
Aconiti Ciliare Tuber extract emerged as a promising botanical agent for hair growth following a screening of 800 natural products. This extract activated β-catenin target gene transcription factors, promoted neural progenitor cell differentiation, elevated ALP activity, and stimulated DP cell proliferation in vitro. It also induced early anagen phase initiation in a mouse model [153].
Nephelium lappaceum var. pallens (Hiern) Leenh. extract demonstrated preventive effects against AA in both in vitro and in vivo testosterone-induced hair loss models. The extract enhanced cell proliferation in the presence of testosterone in HFDPC cultures, and it accelerated hair growth, and upregulated PCNA and cyclin D1 expression in testosterone-induced hair loss model mice [154]. Similarly, Terminalia bellirica (Gaertn.) Roxb. fruit extract was found to counteract the testosterone-induced inhibition of hair growth in C57BL/6 mice. Oral administration restored hair growth markers, including cyclin D1 and PCNA, to near-pretreatment levels and activated Wnt/β-catenin signaling pathways. Furthermore, it increased HF density [155].
Overall, multiple natural products have demonstrated substantial hair growth-promoting effects. However, the clinical relevance of these products remains to be verified and their active compounds should be identified.
Hair Growth-Promoting Phytochemicals and Their Mechanisms of Action
Natural products are composed of a diverse array of components with intricate structures, and the specific bioactive compounds within a given extract determine its physiological effects. Identifying and characterizing compounds that promote hair growth is critical for developing novel therapeutic agents containing active ingredients. Here, we summarize key phytochemicals and their mechanisms of action, presenting a novel framework for advancing hair growth research (Table 4 and Fig. 5).
Table 4.
Mechanisms of action of compounds promoting hair growth.
|
Fig. 5. Known signaling pathways affecting hair health that are targeted by phytochemicals and pharmaceutical compounds.
Tectoridin, an isoflavone derived from the rhizome of the Chinese medicinal herb Belamcanda chinensis (L.) DC., has been shown to activate the Wnt/β-catenin pathway by modulating luciferase activity and enhancing the expression of downstream target genes [156].
Quercitrin (quercetin-3-O-rhamnoside), a natural flavonoid found in various plants, predominantly in the flowers, fruits, and leaves, significantly enhanced cell proliferation in hDPCs, with effects comparable to those of 100 mM minoxidil. Furthermore, quercitrin increased mitochondrial membrane potential, a critical marker of energy metabolism. It also modulated the expression of apoptosis-related genes and proteins, upregulating Bcl2 and Ki67 while downregulating Bad and Bax. Additionally, quercitrin upregulated growth factor expression and phosphorylated AKT, ERK, and CREB-1, thereby activating both receptor tyrosine kinases and non-receptor tyrosine kinases [157].
Morroniside, a glycoside derived from Cornus officinalis, activates Wnt/β-catenin signaling, promoting ORSC proliferation and transition from the telogen to the anagen phase [158].
Autophagy plays a pivotal role in the differentiation and self-renewal of epidermal and dermal stem cells [159], and topical applications of α-ketoglutarate, the AMP-activated protein kinase activator 5-aminoimidazole-4-carboxamide ribonucleotide, metformin, and α-ketobutyrate enhanced hair regeneration by increasing melanin pigmentation, Ki67 expression, and autophagy-related protein expression in mouse skin tissue [160].
A polyphenolic compound 3,4,5-tri-O-caffeoylquinic acid stimulates hair growth via activation of the β-catenin pathway. Topical application of this compound prolonged hair length, an indicator of hair regrowth in mice. Microarray analysis revealed that skin treated with 3,4,5-tri-O-caffeoylquinic acid exhibited differential expression of 1,235 genes, including 435 upregulated and 800 downregulated genes. Of these, the upregulated genes were predominantly associated with the anagen phase and hair growth, while downregulated genes were linked to the telogen phase, apoptosis, cell cycle arrest, and repression of Wnt/β-catenin signaling. Enhanced β-catenin expression was observed in HFs and the epidermis of treated mouse skin. Furthermore, the treatment increased ATP content, ALP activity, and β-catenin expression in HFDPCs [161].
Alpinetin, a major constituent of herbs in the Zingiberaceae and Fabaceae families, has been shown to promote hair growth both in vitro and in vivo. In mice, alpinetin application altered skin pigmentation in mice, increased HF length 7 days post-depilation, and extended HS length at 13 and 17 days after depilation. It accelerated entry into the anagen phase and delayed the transition to the catagen phase by inhibiting cell apoptosis. Alpinetin also activated the expression of hair stem cell markers, and RNA-seq analysis revealed the upregulation of genes involved in cell migration, skin development, hair cycle regulation, and the Wnt signaling pathway. Importantly, alpinetin exhibited no cytotoxic effects in human and mouse primary fibroblasts and keratinocytes [162].
Sinapic acid, a hydroxycinnamic acid found in various herbs, vegetables, high-bran cereals, and fruits, has been proposed as a therapeutic candidate for hair loss. In HFDPCs, sinapic acid enhanced cell proliferation and increased VEGF production. Additionally, it promoted cell cycle progression by facilitating transitions to the S and G2/M phases, an effect mediated by AKT phosphorylation and the subsequent inactivation of the Wnt/β-catenin signaling [163].
Cyanidin 3-O-arabinoside protected DP cells from DHT-induced senescence and mitochondrial dysfunction by reducing mitochondrial ROS and regulating NADPH oxidase (NOX)-mediated MAPK signaling. It also suppressed the phosphorylation of p38 and HSP27 by regulating ROS formation in a NOX-dependent manner. Moreover, the compound inhibited ER–mitochondria contact and mitochondrial calcium accumulation. In vivo, the topical application of cyanidin 3-O-arabinoside accelerated hair growth in an AA-like mouse model [164].
Decursin, a pyranocoumarin compound and the primary bioactive component of Angelica gigas, was examined for its potential therapeutic effects on alopecia. A comprehensive literature review utilizing the PubChem database identified decursin as being closely associated with alopecia and implicated it in the apoptosis pathway related to chemotherapy-induced alopecia. In cyclophosphamide-treated mice, decursin reduced apoptosis markers and suppressed PI3K/AKT and MAPK signaling molecules [165].
Loliolide, a monoterpenoid, demonstrated therapeutic potential for alopecia by promoting the proliferation and 3D-culture formation of HDP cells. Additionally, the loliolide treatment activated the AKT signaling cascade, upregulating Wnt/β-catenin signaling via AKT pathway activation [166] (Fig. 5).
In this review, we systematically organized the known natural phytochemicals that exhibit promising effects on hair growth. Natural products contain a diverse array of bioactive compounds and, thus, exert multifaceted effects, necessitating the identification of key constituents in order to standardize product development for hair health. Despite extensive investigation into the efficacy of these compounds and the mechanisms driving their effects, none have yet translated into clinical applications. Consequently, further research is needed to evaluate phytochemicals with potential hair growth-promoting properties and minimal adverse effects on non-target organs.
Conclusion and Future Research Strategies
Recent advancements in hair growth research have prioritized the development of safe and effective therapeutic materials with minimal adverse effects on non-target organs. A detailed understanding of the mechanisms regulating hair growth and the identification of treatment targets are essential for identifying new therapeutic candidates. In this review, we comprehensively examined the key aspects of the hair growth process, hair follicle morphogenesis and regeneration, the factors regulating follicle regeneration, current anti-hair loss drugs, and the novel hair growth-promoting agents.
Our review highlights the potential of natural product-derived phytochemicals as targeted modulators of molecular pathways involved in hair growth. Such approaches offer a promising avenue for maintaining hair health, accelerating the hair cycle, and promoting hair regeneration. Moreover, we revisited previously studied materials, offering innovative perspectives and suggesting strategies to refine and advance existing research paradigms in this field. These insights contribute to a more comprehensive understanding of the mechanisms promoting hair growth and support the development of effective phytochemical-based therapeutic agents that are both effective and exhibit minimal systemic side effects.
Acknowledgments
This study was financially supported by the Basic Science Research Program through the National Research Foundation of Korea, which is funded by the Ministry of Education (NRF-2022R1A2C1010923).
Footnotes
Conflicts of Interest
The authors have no financial conflicts of interest to declare.
References
- 1.Noback CR. Morphology and phylogeny of hair. Ann. NY Acad. Sci. 1951;53:476–492. doi: 10.1111/j.1749-6632.1951.tb31950.x. [DOI] [PubMed] [Google Scholar]
- 2.Randall VA, Botchkareva NV. 2009. The biology of hair growth. In, Cosmetics Applications of Laser & Light-Based Systems, pp. 3-35.
- 3.Buffoli B, Rinaldi F, Labanca M, Sorbellini E, Trink A, Guanziroli E, et al. The human hair: from anatomy to physiology. Int. J. Dermatol. 2013;53:331–341. doi: 10.1111/ijd.12362. [DOI] [PubMed] [Google Scholar]
- 4.Maffei C. Personality disorders and psychopathologic symptoms in patients with androgenetic alopecia. Arch. Dermatol. 1994;130:868–872. doi: 10.1001/archderm.1994.01690070062009. [DOI] [PubMed] [Google Scholar]
- 5.Mason KA, Schoelwer MJ, Rogol AD. Androgens during infancy, childhood, and adolescence: physiology and use in clinical practice. Endocr. Rev. 2020;41:bnaa003. doi: 10.1210/endrev/bnaa003. [DOI] [PubMed] [Google Scholar]
- 6.Trueb R, Henry J, Davis M, Schwartz J. Scalp condition impacts hair growth and retention via oxidative stress. Int. J. Trichol. 2018;10:262–270. doi: 10.4103/ijt.ijt_57_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kaiser M, Abdin R, Gaumond SI, Issa NT, Jimenez JJ. Treatment of androgenetic alopecia: current guidance and unmet needs. Clin. Cosmet. Investig. Dermatol. 2023;16:1387–1406. doi: 10.2147/CCID.S385861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sattur SS, Sattur IS. Pharmacological management of pattern hair loss. Indian J. Plast. Surg. 2021;54:422–434. doi: 10.1055/s-0041-1739254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kaushik D, Gupta D, Yadav R. ALOPECIA: HERBAL REMEDIES. Int. J. Pharm. Sci. Res. 2011;2:1631–1637. [Google Scholar]
- 10.Ji S, Zhu Z, Sun X, Fu X. Functional hair follicle regeneration: an updated review. Signal Transduct. Target. Ther. 2021;6:66. doi: 10.1038/s41392-020-00441-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Blume-Peytavi U, Vogt A. Human hair follicle: reservoir function and selective targeting. Br. J. Dermatol. 2011;165 Suppl 2:13–17. doi: 10.1111/j.1365-2133.2011.10572.x. [DOI] [PubMed] [Google Scholar]
- 12.Soni RB, Deshmukh J, Arya RS. HUMAN HAIRS-'Why we have long scalp hairs but short body hairs'. IOS.R J. Dental Med.Sci. 2018;17:58–63. [Google Scholar]
- 13.Chen CL, Huang WY, Wang EHC, Tai KY, Lin SJ. Functional complexity of hair follicle stem cell niche and therapeutic targeting of niche dysfunction for hair regeneration. J. Biomed. Sci. 2020;27:43. doi: 10.1186/s12929-020-0624-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vogt A, Hadam S, Heiderhoff M, Audring H, Lademann J, Sterry W, et al. Morphometry of human terminal and vellus hair follicles. Exp. Dermatol. 2007;16:946–950. doi: 10.1111/j.1600-0625.2007.00602.x. [DOI] [PubMed] [Google Scholar]
- 15.Schneider MR, Schmidt-Ullrich R, Paus R. The hair follicle as a dynamic miniorgan. Curr. Biol. 2009;19:R132–142. doi: 10.1016/j.cub.2008.12.005. [DOI] [PubMed] [Google Scholar]
- 16.Stenn KS, Paus R. Controls of hair follicle cycling. Physiol. Rev. 2001;81:449–494. doi: 10.1152/physrev.2001.81.1.449. [DOI] [PubMed] [Google Scholar]
- 17.Rendl M, Lewis L, Fuchs E. Molecular dissection of mesenchymal-epithelial interactions in the hair follicle. PLoS Biol. 2005;3:e331. doi: 10.1371/journal.pbio.0030331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ito M. The innermost cell layer of the outer root sheath in anagen hair follicle: light and electron microscopic study. Arch. Dermatol. Res. 1986;279:112–119. doi: 10.1007/BF00417531. [DOI] [PubMed] [Google Scholar]
- 19.Joshi RS. The inner root sheath and the men associated with it eponymically. Int. J. Trichology. 2011;3:57–62. doi: 10.4103/0974-7753.82119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Harkey MR. Anatomy and physiology of hair. Forensic Sci. Int. 1993;63:9–18. doi: 10.1016/0379-0738(93)90255-9. [DOI] [PubMed] [Google Scholar]
- 21.Wang X, Tredget EE, Wu Y. Dynamic signals for hair follicle development and regeneration. Stem Cells Dev. 2012;21:7–18. doi: 10.1089/scd.2011.0230. [DOI] [PubMed] [Google Scholar]
- 22.Sennett R, Rendl M. Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling. Semin. Cell Dev. Biol. 2012;23:917–927. doi: 10.1016/j.semcdb.2012.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Higgins CA, Westgate GE, Jahoda CA. From telogen to exogen: mechanisms underlying formation and subsequent loss of the hair club fiber. J. Invest. Dermatol. 2009;129:2100–2108. doi: 10.1038/jid.2009.66. [DOI] [PubMed] [Google Scholar]
- 24.Reynolds AJ, Lawrence C, Cserhalmi-Friedman PB, Christiano AM, Jahoda CA. Trans-gender induction of hair follicles. Nature. 1999;402:33–34. doi: 10.1038/46938. [DOI] [PubMed] [Google Scholar]
- 25.van der Veen C, Handjiski B, Paus R, Müller-Röver S, Maurer M, Eichmüller S, et al. A comprehensive guide for the recognition and classification of distinct stages of hair follicle morphogenesis. J. Invest. Dermatol. 1999;113:523–532. doi: 10.1046/j.1523-1747.1999.00740.x. [DOI] [PubMed] [Google Scholar]
- 26.Zhang X, Ye Y, Zhu Z, Yang Y, Cao H, McElwee KJ, et al. Sequential cyclic changes of hair roots revealed by dermoscopy demonstrate a progressive mechanism of diffuse alopecia areata over time. Exp. Dermatol. 2020;29:223–230. doi: 10.1111/exd.13799. [DOI] [PubMed] [Google Scholar]
- 27.Stenn K. 1998. Growth of the hair follicle: a cycling and regenerating biological system. Molecular basis of epithelial appendage morphogenesis.
- 28.Milner Y, Sudnik J, Filippi M, Kizoulis M, Kashgarian M, Stenn K. Exogen, shedding phase of the hair growth cycle: characterization of a mouse model. J. Invest. Dermatol. 2002;119:639–644. doi: 10.1046/j.1523-1747.2002.01842.x. [DOI] [PubMed] [Google Scholar]
- 29.Higgins CA, Richardson GD, Westgate GE, Jahoda CA. Exogen involves gradual release of the hair club fibre in the vibrissa follicle model. Exp. Dermatol. 2009;18:793–795. doi: 10.1111/j.1600-0625.2008.00833.x. [DOI] [PubMed] [Google Scholar]
- 30.Koch PJ, Mahoney MG, Cotsarelis G, Rothenberger K, Lavker RM, Stanley JR. Desmoglein 3 anchors telogen hair in the follicle. J. Cell Sci. 1998;111:2529–2537. doi: 10.1242/jcs.111.17.2529. [DOI] [PubMed] [Google Scholar]
- 31.Lai-Cheong JE, McGrath JA. Structure and function of skin, hair and nails. Medicine. 2021;49:337–342. doi: 10.1016/j.mpmed.2021.03.001. [DOI] [Google Scholar]
- 32.Wang ECE, Higgins CA. Immune cell regulation of the hair cycle. Exp. Dermatol. 2020;29:322–333. doi: 10.1111/exd.14070. [DOI] [PubMed] [Google Scholar]
- 33.Alhilli F, Wright EA. The effects of environmental-temperature on the hair coat of the mouse. J. Thermal Biol. 1988;13:21–24. doi: 10.1016/0306-4565(88)90005-8. [DOI] [Google Scholar]
- 34.Schmidt-Ullrich R, Paus R. Molecular principles of hair follicle induction and morphogenesis. Bioessays. 2005;27:247–261. doi: 10.1002/bies.20184. [DOI] [PubMed] [Google Scholar]
- 35.Kligman AM, Strauss JS. The formation of vellus hair follicles from human adult epidermis. J. Invest. Dermatol. 1956;27:19–23. doi: 10.1038/jid.1956.71. [DOI] [PubMed] [Google Scholar]
- 36.Wier EM, Garza LA. Through the lens of hair follicle neogenesis, a new focus on mechanisms of skin regeneration after wounding. Semin. Cell Dev. Biol. 2020;100:122–129. doi: 10.1016/j.semcdb.2019.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rishikaysh P, Dev K, Diaz D, Qureshi WM, Filip S, Mokry J. Signaling involved in hair follicle morphogenesis and development. Int. J. Mol. Sci. 2014;15:1647–1670. doi: 10.3390/ijms15011647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yang CC, Cotsarelis G. Review of hair follicle dermal cells. J. Dermatol. Sci. 2010;57:2–11. doi: 10.1016/j.jdermsci.2009.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Millar SE. Molecular mechanisms regulating hair follicle development. J. Invest. Dermatol. 2002;118:216–225. doi: 10.1046/j.0022-202x.2001.01670.x. [DOI] [PubMed] [Google Scholar]
- 40.Driskell RR, Clavel C, Rendl M, Watt FM. Hair follicle dermal papilla cells at a glance. J. Cell Sci. 2011;124:1179–1182. doi: 10.1242/jcs.082446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Akiyama M, Smith LT, Shimizu H. Changing patterns of localization of putative stem cells in developing human hair follicles. J. Invest. Dermatol. 2000;114:321–327. doi: 10.1046/j.1523-1747.2000.00857.x. [DOI] [PubMed] [Google Scholar]
- 42.Rebora A, Guarrera M. 2002. Kenogen. A new phase of the hair cycle? Dermatology 205: 108-110. 10.1159/000063908 [DOI] [PubMed]
- 43.Akiyama M, Matsuo I, Shimizu H. Formation of cornified cell envelope in human hair follicle development. Br. J. Dermatol. 2002;146:968–976. doi: 10.1046/j.1365-2133.2002.04869.x. [DOI] [PubMed] [Google Scholar]
- 44.Holbrook KA, Minami SI. Hair follicle embryogenesis in the human. Characterization of events in vivo and in vitro. Ann. N Y Acad. Sci. 1991;642:167–196. doi: 10.1111/j.1749-6632.1991.tb24387.x. [DOI] [PubMed] [Google Scholar]
- 45.Breathnach AS, Robins EJ. 1981. Ultrastructure of developing hair tract and hair canal in the human fetus. In Hair Research, Springer, pp. 12-17. 10.1007/978-3-642-81650-5_2 [DOI]
- 46.Peters EM, Botchkarev VA, Muller-Rover S, Moll I, Rice FL, Paus R. Developmental timing of hair follicle and dorsal skin innervation in mice. J. Comp. Neurol. 2002;448:28–52. doi: 10.1002/cne.10212. [DOI] [PubMed] [Google Scholar]
- 47.Vogt A, McElwee KJ, Blume-Peytavi U. 2008. Biology of the hair follicle. In Whitting DA, Blume-Peytavi U, Tosti A, Trüeb RM (eds.), Hair Growth and Disorders, Springer Berlin Heidelberg, Berlin, Heidelberg, pp. 1-22. 10.1007/978-3-540-46911-7_1 [DOI]
- 48.Hardy MH. 1969. The differentiation of hair follicles and hairs in organ culture. Advances in Biology of Skin. pp. 35-60.
- 49.Houschyar KS, Borrelli MR, Tapking C, Popp D, Puladi B, Ooms M, et al. Molecular mechanisms of hair growth and regeneration: current understanding and novel paradigms. Dermatology. 2020;236:271–280. doi: 10.1159/000506155. [DOI] [PubMed] [Google Scholar]
- 50.Park AM, Khan S, Rawnsley J. Hair biology: growth and pigmentation. Facial Plast Surg. Clin. North Am. 2018;26:415–424. doi: 10.1016/j.fsc.2018.06.003. [DOI] [PubMed] [Google Scholar]
- 51.Stenn KS, Paus R. What controls hair follicle cycling? Exp. Dermatol. 1999;8:229–233. discussion 233–226. doi: 10.1111/j.1600-0625.1999.tb00376.x. [DOI] [PubMed] [Google Scholar]
- 52.Panteleyev AA, Jahoda CA, Christiano AM. Hair follicle predetermination. J. Cell Sci. 2001;114:3419–3431. doi: 10.1242/jcs.114.19.3419. [DOI] [PubMed] [Google Scholar]
- 53.Porter RM, Gandhi M, Wilson NJ, Wood P, McLean WH, Lane EB. Functional analysis of keratin components in the mouse hair follicle inner root sheath. Br. J. Dermatol. 2004;150:195–204. doi: 10.1111/j.1365-2133.2004.05720.x. [DOI] [PubMed] [Google Scholar]
- 54.Muller-Rover S, Handjiski B, van der Veen C, Eichmuller S, Foitzik K, McKay IA, et al. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Invest. Dermatol. 2001;117:3–15. doi: 10.1046/j.0022-202x.2001.01377.x. [DOI] [PubMed] [Google Scholar]
- 55.Oh JW, Kloepper J, Langan EA, Kim Y, Yeo J, Kim MJ, et al. A guide to studying human hair follicle cycling in vivo. J. Invest. Dermatol. 2016;136:34–44. doi: 10.1038/JID.2015.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Commo S, Bernard BA. Immunohistochemical analysis of tissue remodelling during the anagen-catagen transition of the human hair follicle. Br. J. Dermatol. 1997;137:31–38. doi: 10.1111/j.1365-2133.1997.tb03697.x. [DOI] [PubMed] [Google Scholar]
- 57.Kloepper JE, Sugawara K, Al-Nuaimi Y, Gaspar E, van Beek N, Paus R. Methods in hair research: how to objectively distinguish between anagen and catagen in human hair follicle organ culture. Exp. Dermatol. 2010;19:305–312. doi: 10.1111/j.1600-0625.2009.00939.x. [DOI] [PubMed] [Google Scholar]
- 58.Lowry WE, Blanpain C, Nowak JA, Guasch G, Lewis L, Fuchs E. Defining the impact of beta-catenin/Tcf transactivation on epithelial stem cells. Genes Dev. 2005;19:1596–1611. doi: 10.1101/gad.1324905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yang C-C, Sheu H-M, Chung P-L, Chang C-H, Tsai Y-S, Hughes MW, et al. Leptin of dermal adipose tissue is differentially expressed during the hair cycle and contributes to adipocyte-mediated growth inhibition of anagen-phase vibrissa hair. Exper. Dermatol. 2015;24:57–60. doi: 10.1111/exd.12566. [DOI] [PubMed] [Google Scholar]
- 60.Van Neste D, Leroy T, Conil S. Exogen hair characterization in human scalp. Skin Res. Technol. 2007;13:436–443. doi: 10.1111/j.1600-0846.2007.00248.x. [DOI] [PubMed] [Google Scholar]
- 61.Liyanage D, Sinclair R. Telogen Effluvium. Cosmetics. 2016;3:13. doi: 10.3390/cosmetics3020013. [DOI] [Google Scholar]
- 62.Zhou G, Kang D, Ma S, Wang X, Gao Y, Yang Y, et al. Integrative analysis reveals ncRNA-mediated molecular regulatory network driving secondary hair follicle regression in cashmere goats. BMC Genomics. 2018;19:222. doi: 10.1186/s12864-018-4603-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rishikaysh P, Dev K, Diaz D, Qureshi WMS, Filip S, Mokry J. Signaling involved in hair follicle morphogenesis and development. Int. J. Mol. Sci. 2014;15:1647–1670. doi: 10.3390/ijms15011647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang X, Liu Y, He J, Wang J, Chen X, Yang R. Regulation of signaling pathways in hair follicle stem cells. Burns Trauma. 2022;10:tkac022. doi: 10.1093/burnst/tkac022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shimomura Y, Christiano AM. Biology and genetics of hair. Ann. Rev. Genomics Human Genet. 2010;11:109–132. doi: 10.1146/annurev-genom-021610-131501. [DOI] [PubMed] [Google Scholar]
- 66.Shang S, Hua F, Hu ZW. The regulation of beta-catenin activity and function in cancer: therapeutic opportunities. Oncotarget. 2017;8:33972–33989. doi: 10.18632/oncotarget.15687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, et al. Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022;7:1–23. doi: 10.1038/s41392-021-00762-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kwack MH, Kim MK, Kim JC, Sung YK. Dickkopf 1 Promotes Regression of Hair Follicles. J. Invest. Dermatol. 2012;132:1554–1560. doi: 10.1038/jid.2012.24. [DOI] [PubMed] [Google Scholar]
- 69.Kwack MH, Sung YK, Chung EJ, Im SU, Ahn JS, Kim MK, et al. Dihydrotestosterone-inducible dickkopf 1 from balding dermal papilla cells causes apoptosis in follicular keratinocytes. J. Invest. Dermatol. 2008;128:262–269. doi: 10.1038/sj.jid.5700999. [DOI] [PubMed] [Google Scholar]
- 70.Qin K, Yu M, Fan J, Wang H, Zhao P, Zhao G, et al. Canonical and noncanonical Wnt signaling: multilayered mediators, signaling mechanisms and major signaling crosstalk. Genes Dis. 2024;11:103–134. doi: 10.1016/j.gendis.2023.01.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yu M, Qin K, Fan J, Zhao G, Zhao P, Zeng W, et al. The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities. Genes Dis. 2024;11:101026. doi: 10.1016/j.gendis.2023.04.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.St-Jacques B, Dassule HR, Karavanova I, Botchkarev VA, Li J, Danielian PS, et al. Sonic hedgehog signaling is essential for hair development. Curr. Biol. 1998;8:1058–1068. doi: 10.1016/S0960-9822(98)70443-9. [DOI] [PubMed] [Google Scholar]
- 73.Pham A, Therond P, Alves G, Tournier FB, Busson D, Lamourisnard C, et al. The suppressor of fused gene encodes a novel pest protein involved in drosophila segment polarity establishment. Genetics. 1995;140:587–598. doi: 10.1093/genetics/140.2.587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Briscoe J, Thérond PP. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 2013;14:416–429. doi: 10.1038/nrm3598. [DOI] [PubMed] [Google Scholar]
- 75.Severini LL, Quaglio D, Basili I, Ghirga F, Bufalieri F, Caimano M, et al. A Smo/Gli multitarget hedgehog pathway inhibitor impairs tumor growth. Cancers. 2019;11:1518. doi: 10.3390/cancers11101518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Huang DL, Wang YT, Tang JB, Luo SW. Molecular mechanisms of suppressor of fused in regulating the hedgehog signalling pathway (Review) Oncol. Lett. 2018;15:6077–6086. doi: 10.3892/ol.2018.8142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sabol M, Trnski D, Musani V, Ozretic P, Levanat S. Role of GLI transcription factors in pathogenesis and their potential as new therapeutic Targets. Int. J. Mol. Sci. 2018;19:2562. doi: 10.3390/ijms19092562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Abe Y, Tanaka N. Roles of the hedgehog signaling pathway in epidermal and hair follicle development, homeostasis, and cancer. J. Dev. Biol. 2017;5:12. doi: 10.3390/jdb5040012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hu XM, Li ZX, Zhang DY, Yang YC, Fu SA, Zhang ZQ, et al. A systematic summary of survival and death signalling during the life of hair follicle stem cells. Stem Cell Res. Ther. 2021;12:453. doi: 10.1186/s13287-021-02527-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.St-Jacques B, Dassule H, Karavanova I, Botchkarev V, Li J, Danielian P, et al. Sonic hedgehog signaling is essential for hair development. Curr. Biol. 1998;8:1058–1069. doi: 10.1016/S0960-9822(98)70443-9. [DOI] [PubMed] [Google Scholar]
- 81.O'Shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity. 2012;36:542–550. doi: 10.1016/j.immuni.2012.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu. Rev. Med. 2015;66:311–328. doi: 10.1146/annurev-med-051113-024537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gilhar A, Kalish RS. Alopecia areata: a tissue specific autoimmune disease of the hair follicle. Autoimmun. Rev. 2006;5:64–69. doi: 10.1016/j.autrev.2005.07.001. [DOI] [PubMed] [Google Scholar]
- 84.Divito SJ, Kupper TS. Inhibiting Janus kinases to treat alopecia areata. Nat. Med. 2014;20:989–990. doi: 10.1038/nm.3685. [DOI] [PubMed] [Google Scholar]
- 85.Dodington DW, Desai HR, Woo M. JAK/STAT - emerging players in metabolism. Trends Endocrinol. Metab. 2018;29:55–65. doi: 10.1016/j.tem.2017.11.001. [DOI] [PubMed] [Google Scholar]
- 86.Hu XY, Li J, Fu MR, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct. Target. Ther. 2021;6:402. doi: 10.1038/s41392-021-00791-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ha Kim B, Lee WY, Trinh TA, Pyo JS, Lee S, Kim CE, et al. Hair growth effect of emulsion extracted brevilin A, a JAK3 inhibitor, from Centipeda minima. Processes. 2020;8:767. doi: 10.3390/pr8070767. [DOI] [Google Scholar]
- 88.Jones MC. Treatment options for androgenetic alopecia. Us Pharmacist. 2018;43:12–16. [Google Scholar]
- 89.Ramanlal R, Gupta V. 2024. Physiology, vasodilation. In, StatPearls, Treasure Island (FL). [PubMed]
- 90.Murphrey MB, Agarwal S, Zito PM. 2024. Anatomy, hair. In, StatPearls, Treasure Island (FL). [PubMed]
- 91.Ziche M, Morbidelli L. Nitric oxide and angiogenesis. J. Neurooncol. 2000;50:139–148. doi: 10.1023/A:1006431309841. [DOI] [PubMed] [Google Scholar]
- 92.Kwack MH, Kang BM, Kim MK, Kim JC, Sung YK. Minoxidil activates beta-catenin pathway in human dermal papilla cells: a possible explanation for its anagen prolongation effect. J. Dermatol. Sci. 2011;62:154–159. doi: 10.1016/j.jdermsci.2011.01.013. [DOI] [PubMed] [Google Scholar]
- 93.Hsu CL, Liu JS, Lin AC, Yang CH, Chung WH, Wu WG. Minoxidil may suppress androgen receptor-related functions. Oncotarget. 2014;5:2187–2197. doi: 10.18632/oncotarget.1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Han JH, Kwon OS, Chung JH, Cho KH, Eun HC, Kim KH. Effect of minoxidil on proliferation and apoptosis in dermal papilla cells of human hair follicle. J. Dermatol. Sci. 2004;34:91–98. doi: 10.1016/j.jdermsci.2004.01.002. [DOI] [PubMed] [Google Scholar]
- 95.Buhl AE, Waldon DJ, Kawabe TT, Holland JM. Minoxidil stimulates mouse vibrissae follicles in organ culture. J. Invest. Dermatol. 1989;92:315–320. doi: 10.1111/1523-1747.ep12277095. [DOI] [PubMed] [Google Scholar]
- 96.Olsen EA, Dunlap FE, Funicella T, Koperski JA, Swinehart JM, Tschen EH, et al. A randomized clinical trial of 5% topical minoxidil versus 2% topical minoxidil and placebo in the treatment of androgenetic alopecia in men. J. Am. Acad. Dermatol. 2002;47:377–385. doi: 10.1067/mjd.2002.124088. [DOI] [PubMed] [Google Scholar]
- 97.Olsen EA, Whiting D, Bergfeld W, Miller J, Hordinsky M, Wanser R, et al. A multicenter, randomized, placebo-controlled, double-blind clinical trial of a novel formulation of 5% minoxidil topical foam versus placebo in the treatment of androgenetic alopecia in men. J. Am. Acad. Dermatol. 2007;57:767–774. doi: 10.1016/j.jaad.2007.04.012. [DOI] [PubMed] [Google Scholar]
- 98.Blume-Peytavi U, Hillmann K, Dietz E, Canfield D, Garcia Bartels N. 2011. A randomized, single-blind trial of 5% minoxidil foam once daily versus 2% minoxidil solution twice daily in the treatment of androgenetic alopecia in women. J. Am. Acad. Dermatol. 65: 1126-1134 e1122. 10.1016/j.jaad.2010.09.724 [DOI] [PubMed]
- 99.Escamilla-Cruz M, Magana M, Escandon-Perez S, Bello-Chavolla OY. Use of 5-alpha reductase inhibitors in dermatology: a narrative review. Dermatol. Ther. (Heidelb) 2023;13:1721–1731. doi: 10.1007/s13555-023-00974-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Salisbury BH, Tadi P. 2024. 5-Alpha-Reductase Inhibitors. In, StatPearls, Treasure Island (FL).
- 101.Nickel JC. Comparison of clinical trials with finasteride and dutasteride. Rev Urol. 2004;6 Suppl 9:S31–39. [PMC free article] [PubMed] [Google Scholar]
- 102.Rattanachitthawat N, Pinkhien T, Opanasopit P, Ngawhirunpat T, Chanvorachote P. Finasteride enhances stem cell signals of human dermal papilla cells. In Vivo. 2019;33:1209–1220. doi: 10.21873/invivo.11592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Sugar AM, Alsip SG, Galgiani JN, Graybill JR, Dismukes WE, Cloud GA, et al. Pharmacology and toxicity of high-dose ketoconazole. Antimicrob. Agents Chemother. 1987;31:1874–1878. doi: 10.1128/AAC.31.12.1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Coskuner ER, Ozkan B, Culha MG. Sexual problems of men with androgenic alopecia treated with 5-alpha reductase inhibitors. Sex Med Rev. 2019;7:277–282. doi: 10.1016/j.sxmr.2018.07.003. [DOI] [PubMed] [Google Scholar]
- 105.Munster U, Hammer S, Blume-Peytavi U, Schafer-Korting M. Testosterone metabolism in human skin cells in vitro and its interaction with estradiol and dutasteride. Skin Pharmacol. Appl. Skin Physiol. 2003;16:356–366. doi: 10.1159/000072930. [DOI] [PubMed] [Google Scholar]
- 106.Manosroi A, Ruksiriwanich W, Manosroi W, Abe M, Manosroi J. In vivo hair growth promotion activity of gel containing niosomes loaded with the Oryza sativa bran fraction (OSF3) Adv. Sci. Lett. 2012;16:222–228. doi: 10.1166/asl.2012.3657. [DOI] [Google Scholar]
- 107.Grino PB, Griffin JE, Wilson JD. Testosterone at high-concentrations interacts with the human androgen receptor similarly to dihydrotestosterone. Endocrinology. 1990;126:1165–1172. doi: 10.1210/endo-126-2-1165. [DOI] [PubMed] [Google Scholar]
- 108.Davey RA, Grossmann M. Androgen receptor structure, function and biology: from bench to bedside. Clin. Biochem. Rev. 2016;37:3–15. [PMC free article] [PubMed] [Google Scholar]
- 109.He Y, Yin D, Perera M, Kirkovsky L, Stourman N, Li W, et al. Novel nonsteroidal ligands with high binding affinity and potent functional activity for the androgen receptor. Eur. J. Med. Chem. 2002;37:619–634. doi: 10.1016/S0223-5234(02)01335-1. [DOI] [PubMed] [Google Scholar]
- 110.Kelly Y, Blanco A, Tosti A. Androgenetic alopecia: an update of treatment options. Drugs. 2016;76:1349–1364. doi: 10.1007/s40265-016-0629-5. [DOI] [PubMed] [Google Scholar]
- 111.Ferreira-Nunes R, Cunha-Filho M, Gratieri T, Gelfuso GM. Follicular-targeted delivery of spironolactone provided by polymeric nanoparticles. Colloids Surf. B Biointerfaces. 2021;208:112101. doi: 10.1016/j.colsurfb.2021.112101. [DOI] [PubMed] [Google Scholar]
- 112.Kiesewetter F, Arai A, Schell H. Sex hormones and antiandrogens influence in vitro growth of dermal papilla cells and outer root sheath keratinocytes of human hair follicles. J. Invest. Dermatol. 1993;101:98S–105S. doi: 10.1111/1523-1747.ep12363015. [DOI] [PubMed] [Google Scholar]
- 113.Zhao J, Harada N, Okajima K. Dihydrotestosterone inhibits hair growth in mice by inhibiting insulin-like growth factor-I production in dermal papillae. Growth Horm. IGF Res. 2011;21:260–267. doi: 10.1016/j.ghir.2011.07.003. [DOI] [PubMed] [Google Scholar]
- 114.Mohler ML, Sikdar A, Ponnusamy S, Hwang DJ, He Y, Miller DD, et al. An overview of next-generation androgen receptortargeted therapeutics in development for the treatment of prostate cancer. Int. J. Mol. Sci. 2021;22:2124. doi: 10.3390/ijms22042124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sintov A, Serafimovich S, Gilhar A. New topical antiandrogenic formulations can stimulate hair growth in human bald scalp grafted onto mice. Int. J. Pharm. 2000;194:125–134. doi: 10.1016/S0378-5173(99)00359-2. [DOI] [PubMed] [Google Scholar]
- 116.Goldspiel BR, Kohler DR. Flutamide - an antiandrogen for advanced prostate-cancer. Dicp Ann. Pharmacother. 1990;24:616–623. doi: 10.1177/106002809002400612. [DOI] [PubMed] [Google Scholar]
- 117.Shin DW. The physiological and pharmacological roles of prostaglandins in hair growth. Korean J. Physiol. Pharmacol. 2022;26:405–413. doi: 10.4196/kjpp.2022.26.6.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Johnstone MA. Hypertrichosis and increased pigmentation of eyelashes and adjacent hair in the region of the ipsilateral eyelids of patients treated with unilateral topical latanoprost. Am. J. Ophthalmol. 1997;124:544–547. doi: 10.1016/S0002-9394(14)70870-0. [DOI] [PubMed] [Google Scholar]
- 119.Sasaki S, Hozumi Y, Kondo S. Influence of prostaglandin F2alpha and its analogues on hair regrowth and follicular melanogenesis in a murine model. Exp. Dermatol. 2005;14:323–328. doi: 10.1111/j.0906-6705.2005.00270.x. [DOI] [PubMed] [Google Scholar]
- 120.Dutkiewicz R, Albert DM, Levin LA. Effects of latanoprost on tyrosinase activity and mitotic index of cultured melanoma lines. Exp. Eye Res. 2000;70:563–569. doi: 10.1006/exer.1999.0819. [DOI] [PubMed] [Google Scholar]
- 121.Oliveira PM, Alencar-Silva T, Pires FQ, Cunha-Filho M, Gratieri T, Carvalho JL, et al. Nanostructured lipid carriers loaded with an association of minoxidil and latanoprost for targeted topical therapy of alopecia. Eur. J. Pharm. Biopharm. 2022;172:78–88. doi: 10.1016/j.ejpb.2022.02.003. [DOI] [PubMed] [Google Scholar]
- 122.Blume-Peytavi U, Lonnfors S, Hillmann K, Garcia Bartels N. 2012. A randomized double-blind placebo-controlled pilot study to assess the efficacy of a 24-week topical treatment by latanoprost 0.1% on hair growth and pigmentation in healthy volunteers with androgenetic alopecia. J. Am. Acad. Dermatol. 66: 794-800. 10.1016/j.jaad.2011.05.026 [DOI] [PubMed]
- 123.Khidhir KG, Woodward DF, Farjo NP, Farjo BK, Tang ES, Wang JW, et al. The prostamide-related glaucoma therapy, bimatoprost, offers a novel approach for treating scalp alopecias. FASEB J. 2013;27:557–567. doi: 10.1096/fj.12-218156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Elias MJ, Weiss J, Weiss E. 2011. Bimatoprost ophthalmic solution 0.03% for eyebrow growth. Dermatol. Surg. 37: 1057-1059. 10.1111/j.1524-4725.2011.01908.x [DOI] [PubMed]
- 125.Rani N, Sharma A, Gupta GK, Singh R. Imidazoles as potential antifungal agents: a review. Mini Rev. Med. Chem. 2013;13:1626–1655. doi: 10.2174/13895575113139990069. [DOI] [PubMed] [Google Scholar]
- 126.Hugo Perez BS. Ketocazole as an adjunct to finasteride in the treatment of androgenetic alopecia in men. Med. Hypotheses. 2004;62:112–115. doi: 10.1016/S0306-9877(03)00264-0. [DOI] [PubMed] [Google Scholar]
- 127.Li B, Mackay DR, Dai Q, Li TW, Nair M, Fallahi M, et al. The LEF1/beta -catenin complex activates movo1, a mouse homolog of Drosophila ovo required for epidermal appendage differentiation. Proc. Natl. Acad. Sci. USA. 2002;99:6064–6069. doi: 10.1073/pnas.092137099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ito T, Tsuji G, Ohno F, Uchi H, Nakahara T, Hashimoto-Hachiya A, et al. Activation of the OVOL1-OVOL2 axis in the hair bulb and in pilomatricoma. Am. J. Pathol. 2016;186:1036–1043. doi: 10.1016/j.ajpath.2015.12.013. [DOI] [PubMed] [Google Scholar]
- 129.Aldhalimi MA, Hadi NR, Ghafil FA. Promotive effect of topical ketoconazole, minoxidil, and minoxidil with tretinoin on hair growth in male mice. ISRN Pharmacol. 2014;2014:575423. doi: 10.1155/2014/575423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Samadi A, Nasrollahi SA, Hashemi A, Kashani MN, Firooz A. Janus kinase (JAK) inhibitors for the treatment of skin and hair disorders: a review of literature. J. Dermatol. Treat. 2017;28:476–483. doi: 10.1080/09546634.2016.1277179. [DOI] [PubMed] [Google Scholar]
- 131.Blair HA. Ritlecitinib: first approval. Drugs. 2023;83:1315–1321. doi: 10.1007/s40265-023-01928-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Harel S, Higgins CA, Cerise JE, Dai Z, Chen JC, Clynes R, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Sci. Adv. 2015;1:e1500973. doi: 10.1126/sciadv.1500973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kim YE, Choi HC, Nam G, Choi BY. Costunolide promotes the proliferation of human hair follicle dermal papilla cells and induces hair growth in C57BL/6 mice. J. Cosmet. Dermatol. 2019;18:414–421. doi: 10.1111/jocd.12674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kim JE, Lee YJ, Park HR, Lee DG, Jeong KH, Kang H. The effect of JAK inhibitor on the survival, anagen re-entry, and hair follicle immune privilege restoration in human dermal papilla cells. Int. J. Mol. Sci. 2020;21:5137. doi: 10.3390/ijms21145137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Jabbari A, Dai ZP, Xing LZ, Cerise JE, Ramot Y, Berkun Y, et al. Reversal of alopecia areata following treatment with the JAK1/2 inhibitor baricitinib. Ebiomedicine. 2015;2:351–355. doi: 10.1016/j.ebiom.2015.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Triyangkulsri K, Suchonwanit P. Role of janus kinase inhibitors in the treatment of alopecia areata. Drug Des. Dev. Ther. 2018;12:2323–2335. doi: 10.2147/DDDT.S172638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ismail FF, Sinclair R. JAK inhibition in the treatment of alopecia areata - a promising new dawn? Expert Rev. Clin. Pharmacol. 2020;13:43–51. doi: 10.1080/17512433.2020.1702878. [DOI] [PubMed] [Google Scholar]
- 138.King B, Guttman-Yassky E, Peeva E, Banerjee A, Sinclair R, Pavel AB, et al. A phase 2a randomized, placebo-controlled study to evaluate the efficacy and safety of the oral Janus kinase inhibitors ritlecitinib and brepocitinib in alopecia areata: 24-week results. J. Am. Acad. Dermatol. 2021;85:379–387. doi: 10.1016/j.jaad.2021.03.050. [DOI] [PubMed] [Google Scholar]
- 139.Dai ZP, Chen J, Chang YQ, Christiano AM. Selective inhibition of JAK3 signaling is sufficient to reverse alopecia areata. JCI Insight. 2021;6:e142205. doi: 10.1172/jci.insight.142205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Papukashvili D, Rcheulishvili N, Liu C, Xie FF, Tyagi D, He YJ, et al. Perspectives on miRNAs targeting DKK1 for developing hair regeneration therapy. Cells. 2021;10:2957. doi: 10.3390/cells10112957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Tang YL, Wang C, Desamero MJM, Kok MK, Chambers JK, Uchida K, et al. The Philippines stingless bee propolis promotes hair growth through activation of Wnt/β-catenin signaling pathway. Exper. Anim. 2023;72:132–139. doi: 10.1538/expanim.22-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Liu X, Ji T, Hu H, Lv X, Zhu G. The hair growth-promoting effect of gardenia florida fruit extract and its molecular regulation. Evid. Based Complement. Alternat. Med. 2022;2022:8498974. doi: 10.1155/2022/8498974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Jung H, Jung DM, Lee SS, Kim EM, Yoon K, Kim KK. Mangifera Indica leaf extracts promote hair growth via activation of Wnt signaling pathway in human dermal papilla cells. Anim. Cells Syst. 2022;26:129–136. doi: 10.1080/19768354.2022.2085790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wang J, Shen H, Chen T, Ma L. Hair growth-promoting effects of Camellia seed cake extract in human dermal papilla cells and C57BL/6 mice. J. Cosmet. Dermatol. 2022;21:5018–5025. doi: 10.1111/jocd.14955. [DOI] [PubMed] [Google Scholar]
- 145.Ramadhani FJ, Bak DH, Kang SH, Park CH, Park SH, Chung BY, et al. The effects of centipedegrass extract on hair growth via promotion of anagen inductive activity. PLoS One. 2022;17:e0265532. doi: 10.1371/journal.pone.0265532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ryu HS, Jeong J, Lee CM, Lee KS, Lee JN, Park SM, et al. Activation of hair cell growth factors by linoleic acid in malva verticillata seed. Molecules. 2021;26:2117. doi: 10.3390/molecules26082117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jin GR, Zhang YL, Yap J, Boisvert WA, Lee BH. Hair growth potential of Salvia plebeia extract and its associated mechanisms. Pharm. Biol. 2020;58:400–409. doi: 10.1080/13880209.2020.1759654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Shin H, Yoo HG, Inui S, Itami S, Kim IG, Cho AR, et al. Induction of transforming growth factor-beta 1 by androgen is mediated by reactive oxygen species in hair follicle dermal papilla cells. BMB Rep. 2013;46:460–464. doi: 10.5483/BMBRep.2013.46.9.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Choi YM, An S, Lee J, Lee JH, Lee JN, Kim YS, et al. Titrated extract of Centella asiatica increases hair inductive property through inhibition of STAT signaling pathway in three-dimensional spheroid cultured human dermal papilla cells. Biosci. Biotechnol. Biochem. 2017;81:2323–2329. doi: 10.1080/09168451.2017.1385383. [DOI] [PubMed] [Google Scholar]
- 150.Jeong GH, Boisvert WA, Xi MZ, Zhang YL, Choi YB, Cho S, et al. Effect of Miscanthus sinensis var. purpurascens flower extract on proliferation and molecular regulation in human dermal papilla cells and stressed C57BL/6 mice. Chinese J. Integr. Med. 2018;24:591–599. doi: 10.1007/s11655-017-2755-7. [DOI] [PubMed] [Google Scholar]
- 151.Kim MH, Choi YY, Cho IH, Hong J, Kim SH, Yang WM. Angelica sinensis induces hair regrowth via the inhibition of apoptosis signaling. Am. J. Chin. Med. 2014;42:1021–1034. doi: 10.1142/S0192415X14500645. [DOI] [PubMed] [Google Scholar]
- 152.Shin HS, Lee JM, Park SY, Yang JE, Kim JH, Yi TH. Hair growth activity of Crataegus pinnatifida on C57BL/6 mouse model. Phytother. Res. 2013;27:1352–1357. doi: 10.1002/ptr.4870. [DOI] [PubMed] [Google Scholar]
- 153.Park PJ, Moon BS, Lee SH, Kim SN, Kim AR, Kim HJ, et al. Hair growth-promoting effect of Aconiti Ciliare Tuber extract mediated by the activation of Wnt/β-catenin signaling. Life Sci. 2012;91:935–943. doi: 10.1016/j.lfs.2012.09.008. [DOI] [PubMed] [Google Scholar]
- 154.Kang HY, Woo MJ, Paik SJ, Choi HJ, Bach TT, Quang BH, et al. Recovery effects of Nephelium lappaceum var. pallens (Hiern) Leenh. extract on testosterone-induced inhibition of hair growth in C57BL/6 mice and human follicular dermal papilla cells. J. Med. Food. 2024;27:167–175. doi: 10.1089/jmf.2023.K.0124. [DOI] [PubMed] [Google Scholar]
- 155.Woo MJ, Kang H, Paik SJ, Choi HJ, Uddin S, Lee S, et al. The in vivo and in vitro effects of Terminalia bellirica (Gaertn.) Roxb. fruit extract on testosterone-induced hair loss. J. Microbiol. Biotechnol. 2023;33:1467–1474. doi: 10.4014/jmb.2306.06004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Yuen GK, Ho BS, Lin LS, Dong TT, Tsim KW. Tectoridin stimulates the activity of human dermal papilla cells and promotes hair shaft elongation in mouse vibrissae hair follicle culture. Molecules. 2022;27:400. doi: 10.3390/molecules27020400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Kim J, Kim SR, Choi YH, Shin JY, Kim CD, Kang NG, et al. Quercitrin stimulates hair growth with enhanced expression of growth factors via activation of MAPK/CREB signaling pathway. Molecules. 2020;25:4004. doi: 10.3390/molecules25174004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Zhou L, Wang H, Jing J, Yu L, Wu X, Lu Z. Morroniside regulates hair growth and cycle transition via activation of the Wnt/beta-catenin signaling pathway. Sci. Rep. 2018;8:13785. doi: 10.1038/s41598-018-32138-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Salemi S, Yousefi S, Constantinescu MA, Fey MF, Simon HU. Autophagy is required for self-renewal and differentiation of adult human stem cells. Cell Res. 2012;22:432–435. doi: 10.1038/cr.2011.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Chai M, Jiang MS, Vergnes L, Fu XD, de Barros SC, Doan NB, et al. Stimulation of hair growth by small molecules that activate autophagy. Cell Rep. 2019;27:3413–3421.e3. doi: 10.1016/j.celrep.2019.05.070. [DOI] [PubMed] [Google Scholar]
- 161.Bejaoui M, Villareal MO, Isoda H. beta-catenin-mediated hair growth induction effect of 3,4,5-tri-O-caffeoylquinic acid. Aging (Albany NY) 2019;11:4216–4237. doi: 10.18632/aging.102048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Fan X, Chen J, Zhang Y, Wang S, Zhong W, Yuan H, et al. Alpinetin promotes hair regeneration via activating hair follicle stem cells. Chin. Med. 2022;17:63. doi: 10.1186/s13020-022-00619-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Woo H, Lee S, Kim S, Park D, Jung E. Effect of sinapic acid on hair growth promoting in human hair follicle dermal papilla cells via Akt activation. Arch. Dermatol. Res. 2017;309:381–388. doi: 10.1007/s00403-017-1732-5. [DOI] [PubMed] [Google Scholar]
- 164.Jung YH, Chae CW, Choi GE, Shin HC, Lim JR, Chang HS, et al. Cyanidin 3-O-arabinoside suppresses DHT-induced dermal papilla cell senescence by modulating p38-dependent ER-mitochondria contacts. J. Biomed. Sci. 2022;29:17. doi: 10.1186/s12929-022-00800-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Kim MH, Park SJ, Yang WM. Network pharmacology study and experimental confirmation revealing the ameliorative effects of decursin on chemotherapy-induced alopecia. Pharmaceuticals. 2021;14:1150. doi: 10.3390/ph14111150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Lee YR, Bae S, Kim JY, Lee J, Cho DH, Kim HS, et al. Monoterpenoid loliolide regulates hair follicle inductivity of human dermal papilla cells by activating the AKT/β-catenin signaling pathway. J. Microbiol. Biotechnol. 2019;29:1830–1840. doi: 10.4014/jmb.1908.08018. [DOI] [PubMed] [Google Scholar]





