- Review
- Open access
- Published:
Progress on mitochondria and hair follicle development in androgenetic alopecia: relationships and therapeutic perspectives
Stem Cell Research & Therapy volume 16, Article number: 44 (2025)
Abstract
Hair loss has long been a significant concern for many individuals. Recent studies have indicated that mitochondria play a more crucial role in hair loss than previously recognized. This review summarizes the connection between mitochondrial dysfunction and hair follicle development, outlines the links between diseases related to mitochondrial disorders and hair issues, and highlights the influence of mitochondrial dysfunction on androgenetic alopecia. We discuss the cellular and signaling mechanisms associated with hair loss and examine how mitochondrial dysfunction, such as insufficient energy supply, signaling irregularities, protein/gene abnormalities, and programmed cell death, can hinder the normal proliferation, differentiation, and growth of hair follicle cells. Furthermore, we discuss current treatment approaches and potential innovative therapies, including mitochondrion-targeting drugs and advanced techniques that directly target hair follicle cells, providing fresh insights into the crucial role of mitochondria in maintaining hair follicle health and managing hair disorders. Furthermore, this review explores future therapeutic strategies and proposes that mitochondrial research could lead to groundbreaking treatments for hair loss, thus providing optimism and new avenues for the treatment of individuals experiencing hair loss. This review not only underscores the central importance of mitochondria in hair health but also emphasizes the importance of advancing research and treatment in this field.
Graphical abstract

Introduction
Hair loss is a widespread issue affecting up to 80% of men and 40% of women globally [1]. It seriously affects an individual's self-esteem and social interactions [2], with androgenetic alopecia (AGA) being the most common type of hair loss [1]. Hair loss usually results from dysfunction of hair follicles, which is influenced by androgen levels and follicular sensitivity to these hormones, causing follicle shrinkage and shortened hair life cycles. In addition, mitochondrial dysfunction plays an important role in hair loss [3]. Mitochondria are responsible not only for energy production but also for various cellular activities, including cell death and differentiation [4]. Previous studies have associated mitochondrial diseases with skin disorders and hair abnormalities, with rashes and hair issues being common symptoms [5]. A French study categorized skin manifestations in pediatric patients with mitochondrial diseases into four main groups: hair abnormalities, rash and pigmentation disorders, hirsutism, and violet extremities [6]. Research has indicated that in patients with AGA, dihydrotestosterone (DHT) not only directly impacts hair follicles but also exacerbates hair follicle cell dysfunction by inducing oxidative stress in mitochondria [7]. This discovery offers a fresh perspective on understanding AGA and suggests that addressing this condition may involve focusing on maintaining mitochondrial health.
Hair follicles
Hair follicles are essential structures in skin development, and they undergo cycles of renewal comprising the anagen, catagen, and telogen phases [8]. The anagen phase determines the length of the hair shaft, primarily through the action of the inner root sheath (IRS) and the hair shaft. The catagen phase is characterized by a significant reduction in the cell cycle due to increased apoptosis in hair follicle cells. This phase concludes when the hair bulb assumes a club-like shape, indicating the termination of active hair growth. The catagen phase subsequently transitions into the telogen phase, a period of rest during which the follicle remains inactive until it is reactivated to initiate a new anagen phase [9]. Various hair loss disorders of clinical significance stem from the early termination of the active anagen phase [10]. Various factors, both internal and external, can impact hair follicle development, including mitochondrial dysfunction [11]. Key signaling pathways, such as the Wnt/β-catenin and bone morphogenetic protein (BMP) pathways, regulate the hair cycle, with Wnt/β-catenin activation promoting growth by supporting stromal cell proliferation, whereas TGF-β/BMP activation inhibits growth by triggering apoptosis in the catagen phase [12].
The hair follicle is anatomically divided into three parts: the funnel, the isthmus, and the bulb. The cells at the base of the follicle produce new hairs via division and differentiation, whereas the dermal papilla (DP) of the bulb promotes hair growth. Structurally, the hair follicle consists of an IRS and an outer root sheath (ORS). The ORS connects epidermal basal cells and serves as a reservoir for hair follicle stem cells (HFSCs). The activation of HFSCs is controlled by signaling molecules released from the DP located underneath the bulge. Factors released from the DP regulate the state of stem cells, and when a critical concentration is reached, HFSCs begin to enter the anagen phase [13].
HFSCs
Hair cycle regulation is governed primarily by HFSCs, which have high potential for differentiation [14]. HFSCs play a key role in hair follicle regeneration, skin healing, maintenance of hair follicle structure and cyclic activity, response to external stimuli, and promotion of wound healing [15]. This process involves multiple signaling pathways, including the Wnt/β-catenin, Hedgehog (Hh), Notch, BMP, and apoptotic signaling pathways [16].
Wnt signaling is essential for the differentiation and proliferation of HFSCs and influences the hair follicle cycle [17]. Hh signaling enhances the activation pathway for HFSC development, although its precise role requires clarification [2]. Regulatory T cells (Tregs) can promote the proliferation and differentiation of HFSCs by expressing the Notch ligand Jagged-1 (Jag1), thereby facilitating hair follicle regeneration [18]. BMP signaling is critical for maintaining the quiescent state of HFSCs during self-renewal and preserving the cellular properties necessary for the generation of transiently expanded cells (TECs). Attenuation or inhibition of BMP signaling activates HFSCs, as observed in the abnormal activation of quiescent HFSCs in mice lacking Bmpr1a expression [2].
Dermal papilla cells
Dermal papilla cells (DPCs) play a key role in regulating hair growth and development by influencing HFSCs through secreted factors that initiate the anagen phase of hair follicles [19]. However, as DPCs age, they hinder the renewal and differentiation of HFSCs, ultimately leading to hair loss [20]. The activation of Wnt signaling is vital for promoting hair growth [21], whereas the activation of ERK in the phosphoinositide 3-kinase/AKT (PI3K/AKT) and MAPK pathways stimulates DPC proliferation [22]. The versican (VCAN) gene has been identified as a key player in hair follicle development, with increased expression of VCAN correlated with improved hair follicle formation [22,23,24].
HFSCs and DPCs are vital in hair follicles and ensure optimal conditions for hair regeneration and growth [21]. The aforementioned signaling pathways are discussed in more detail in the following sections.
Hair follicle keratinocytes
The keratinocytes of the ORS are responsible for producing essential growth factors that control the hair follicle cycle [25]. Specifically, insulin-like growth factor 1 (IGF1) expression increases anagen, whereas TGFβ1 and TGFβ2 expression initiates catagen. ORS keratinocytes not only are equipped with highly functional mitochondria and other skin cells but also have the capacity to produce different keratin proteins [19].
Overall, ORS keratinocytes represent a cell population with significant potential and may serve as a secondary major regulatory center of hair follicle activity in addition to the DP.
Other cells
Matrix cells in the hair bulb exhibit a high mitotic rate and act as the primary proliferative cells during the anagen phase. Melanocytes, located in the inner layers of these cells within the matrix region of the hair follicle, are responsible for melanin production, determining its color [17, 19]. Fibroblasts (including specialized types such as DPCs) provide support and nourishment to the connective tissue sheath near hair follicles, whereas immune cells such as macrophages and Langerhans cells participate in immune surveillance. The adipocytes surrounding hair follicles play a significant role in their growth cycle [19]. The vascular cells surrounding hair follicles supply essential nutrients and oxygen for healthy development.
Signaling pathways/molecules involved in hair follicle development
Wnt/β-catenin signaling
Wnt/β-catenin signaling is essential for hair follicle growth and influences cell differentiation and the hair cycle, particularly during the transition between the anagen and telogen phases [14, 26]. Wnt signaling promotes the accumulation of β-catenin, which translocates to the nucleus to regulate gene transcription and drive hair follicle cell differentiation. The level of β-catenin affects HFSC differentiation and overall follicle development [27]. Abnormal β-catenin expression can lead to structural hair follicle issues or hair loss [28]. WNT10b, a key activator of this pathway, is crucial for the transition of hair follicles into the anagen phase [29, 30], whereas inhibitors such as DKK1 and sFRP1 regulate this process to prevent excessive HFSC proliferation [2].
BMP signaling
BMP signaling, which is initiated through the phosphorylation of BMP and subsequent receptor interactions, plays a crucial role in the regulation of HFSC proliferation and differentiation through the involvement of Smad proteins [2]. Smad proteins, particularly Smad4, play a central role in transmitting TGF-β signals from the cytoplasm to the nucleus, which is essential for hair follicle development and differentiation [31, 32]. BMP-2 and BMP-4 are key to hair follicle development: BMP-2 promotes hair shaft formation, whereas BMP-4 affects follicle growth and HFSC differentiation [33]. BMP-2 expression increases during the telogen phase, whereas BMP-4 expression peaks during catagen, indicating its role in hair cycle regulation [34]. BMP and Wnt signaling cooperate in maintaining HFSCs, with DKK3 inhibiting Wnt signaling in a feedback loop with BMP-4 [14, 35,36,37]. Additionally, miR-29a/b1 modulates BMP signaling by downregulating Bmpr1a, influencing the hair growth cycle [38].
Notch signaling
Notch signaling is integral to the regulation of hair follicle formation and re-epithelialization. Its evolutionary conservation is particularly significant during the advanced stages of embryonic hair follicle development [39, 40]. In mammals, Notch signaling involves four receptors (Notch 1–4) and five ligands (Delta-like 1 [Dll-1, 3, 4], Jagged-1, and Jagged-2), which interact through cell‒cell binding [41]. Upon binding, the Notch receptor undergoes cleavage by γ-secretase, resulting in the release of the Notch intracellular domain (NICD). The NICD subsequently translocates to the nucleus, where it binds to DNA, initiates gene transcription, and activates downstream responses of Wnt/β-catenin signaling, leading to various biological effects [42].
Hh signaling
Hh signaling begins when Shh binds to its receptor Patched (PTCH), activating Smoothened (Smo) and allowing Gli proteins to enter the nucleus, triggering target gene transcription [43, 44]. This pathway supports hair follicle formation, repair, and stem cell maintenance [14]. Hh signaling stimulates dormant HFSC proliferation and plays a role in the regeneration of hair follicles [45]. However, abnormal activation, such as reduced PTCH expression in basal cell carcinoma, can disrupt normal hair growth [46]. Additionally, Hh signaling interacts with Wnt/β-catenin to promote hair follicle maturation during embryonic development [47].
PI3K/AKT signaling
PI3K/AKT signaling is crucial for hair follicle growth, particularly during the transition from the telogen phase to the anagen phase. AKT, a pivotal component in cell survival, inhibits pro-apoptotic proteins, thereby protecting cells from apoptosis [48]. Furthermore, the signaling pathway also promotes the proliferation of HFSCs and interfollicular epidermal stem cells [49,50,51]. Additionally, it contributes to hair follicle regeneration and wound healing processes by initiating the release of cytokines and growth factors [52]. Moreover, the interaction between long noncoding RNA and miR-21 has been shown to enhance the activation of PI3K/AKT signaling [53].
RAS/MAPK signaling
RAS/MAPK signaling involves key kinases (ERK, JNK, p38) that transmit signals from the cell surface to the nucleus, influencing gene expression through phosphorylation [12]. This pathway regulates the hair cycle and HFSC self-renewal, with GAB1 playing a crucial role in controlling these processes [12, 54]. Additionally, RAS/MAPK activation can affect other pathways, such as the inhibition of Shh expression, highlighting its complex role in hair follicle dynamics and maintenance [54]. (Fig. 1 depicts the signaling pathways that play crucial roles in the growth and development of hair follicles.)
Wnt/β-catenin signaling is recognized for its crucial role in regulating HFSCs, DPCs, and keratinocytes. Additionally, the Hh, Notch, and BMP signaling pathways also contribute to the control of HFSCs. Moreover, the PI3K/AKT and RAS/MAPK signaling pathways are predominantly active in DPCs. This depiction highlights the complex interactions and connections between various signaling pathways that impact the destinies and functions of hair follicle cells.
Mitochondrial structure and function
Mitochondria are essential for regulating intracellular and extracellular signaling pathways, acting as central hubs in these processes. They significantly impact cell survival, fate, differentiation, and growth.
Mitochondrial structure
Mitochondria consist of two distinct membranes: an outer membrane that selectively filters small molecules and an inner membrane characterized by invaginations known as cristae, which increase the surface area to facilitate various biochemical reactions [55]. The inner membrane exhibits selective permeability, allowing the passage of molecules such as oxygen, carbon dioxide, and water. Furthermore, it plays a critical role in the transport of proteins involved in the transfer of adenosine triphosphate (ATP) and pyruvate between the cytoplasm and the mitochondria [56]. The region enclosed by the inner membrane, known as the mitochondrial matrix, contains enzymes and mitochondrial DNA (mtDNA), both of which are essential for energy production and the overall functionality of the cell. These specialized structural features enable mitochondria to regulate critical biological processes, including ATP synthesis, calcium homeostasis, and apoptosis [57,58,59].
Mitochondrial function
ATP synthesis
ATP, recognized as the primary unit of cellular energy, is synthesized through oxidative phosphorylation, which occurs in the inner membrane of the mitochondria—organelles responsible for energy production. This process involves specialized protein complexes that facilitate the conversion of nutrients, including glucose and fatty acids, into bioavailable energy [56]. Mutations in mitochondrial DNA (mtDNA) and the intrinsic aging process can compromise cellular energy metabolism by disrupting mitochondrial function, which in turn reduces ATP production [55].
Mitochondrial ROS and oxidative stress
Reactive oxygen species (ROS) are produced as byproducts of mitochondrial respiration and serve dual functions in both cellular signaling and potential damage [3, 60]. Cells maintain a critical balance between the production and removal of ROS to ensure their health. When this balance is disrupted, the accumulation of ROS can exceed the capacity of cellular antioxidant defenses, resulting in oxidative stress and subsequent cellular dysfunction [3, 61,62,63]. In metabolically active cells, abnormalities in mitochondrial proteins can lead to increased oxidative stress and disrupt cellular function, which can significantly impact hair growth [8, 64].
Mitochondrial apoptosis
Mitochondria not only serve as energy-producing organelles of the cell but play crucial roles in regulating the cellular life cycle. During programmed cell death, mitochondria release specific signaling molecules such as cytochrome c, which subsequently activate certain proteases known as caspases to initiate cell death [65,66,67,68]. Additionally, mitochondrion-specific proteins, such as apoptosis-inducing factor (AIF), can promote cell death through pathways that do not depend on caspase activation [69].
Mitochondrial fusion, division, and mitophagy
Mitochondria sustain their function through continuous processes of fusion and fission, working like a dynamic repair system. Fusion enables damaged mitochondria to merge with healthy mitochondria to improve energy production efficiency. This process is regulated by proteins such as MFN1 and OPA1 [70, 71]. Fission, regulated by DRP1, facilitates the even distribution of mitochondria and enables the identification of damaged regions for removal. Under the influence of PINK1 and PARKIN, damaged mitochondria subsequently undergo degradation through a process known as mitophagy, which is crucial for maintaining cellular health [72, 73].
Mitochondria and hair follicles
Mitochondria are critical to the growth and function of hair follicles [74]. Keratinocytes in the human epidermis and hair follicles contain the highest concentrations of mitochondria [74]. Studies have established a strong connection between mitochondrial dysfunction and hair disorders [5]. Additionally, mitochondria are responsible for regulating signaling pathways in the hair follicle cycle. Expanding on the specific signaling mechanisms of hair follicle development mentioned earlier, the following section offers a comprehensive explanation of how mitochondria impact these signaling pathways in hair follicle development.
Signaling pathways and mitochondria in hair follicles
Mitochondrial regulation of Wnt-β-catenin signaling
Mitochondria are crucial regulators of signaling pathways that drive hair follicle development, with mitochondrial ROS playing a key role in the Wnt-β-catenin signaling pathway. In mitochondrial transcription factor A (TFAM)-deficient mice, keratinocytes fail to generate mitochondrial ROS, which disrupts the transmission of the Wnt-β-catenin signals essential for hair follicle development [75]. The absence of mitochondrial ROS leads to impaired Wnt signaling, reducing β-catenin levels and resulting in premature catagen, a phase characterized by hair follicle regression [3].
Mitochondrial regulation of Notch signaling
Mitochondria are integral to the regulation of the Notch signaling pathway, which plays a crucial role in keratinocyte differentiation and hair follicle development. Studies in Drosophila follicle cells have shown that the downregulation of Dynamin-related protein-1 (DRP1), a protein involved in mitochondrial fission, leads to hyperfused mitochondria and inactivates Notch signaling [76]. This, in turn, inhibits follicle cell differentiation but maintains cell proliferation. The mechanism by which mitochondrial shape affects Notch signaling remains unclear, although recent evidence suggests that mitochondrial morphology may influence Notch activity during cellular differentiation processes [3].
In TFAM-deficient mouse models, the absence of mitochondrial ROS disrupts the transduction of Notch signals, impairing both epidermal and hair follicle differentiation [75]. Specifically, TFAM-deficient keratinocytes exhibit reduced expression of Notch target genes, including Hes1, Hey1, and Hey2, which are essential for keratinocyte differentiation. The lack of mitochondrial ROS generation also prevents the activation of key transcripts involved in epidermal structure, such as KRT1 and KRT10, further supporting the role of Notch signaling in hair follicle homeostasis [77].
Mitochondrial regulation of MAPK-ERK-Mfn2 signaling
Sirtuin 1 (SIRT1), an NAD-dependent deacetylase, plays a key role in maintaining mitochondrial homeostasis and protecting HFSCs from inflammation-induced damage [78].
The protective effects of Sirt1 in HFSCs are mediated through the MAPK-ERK-Mfn2 signaling axis. Activation of this pathway is crucial for preserving mitochondrial function, as inhibition of the MAPK-ERK-Mfn2 pathway negates the ability of Sirt1 to promote HFSC survival, mobility, and proliferation under inflammatory conditions. In TNFα-treated HFSCs, Sirt1 overexpression restored mitochondrial respiratory complex expression and improved glucose uptake and lactic acid production, further supporting mitochondrial metabolism. This pathway highlights the critical role of mitochondrial dynamics in stem cell maintenance, especially in the context of inflammatory stress [75, 78].
Mitochondrial regulation of TGF-β/Smad signaling
Studies have shown that key molecules involved in the apoptotic process, such as the anti-apoptotic protein Bcl-2 and the pro-apoptotic proteins Bax, TGF-β2, and cleaved caspase 3, play crucial roles in hair loss. Inhibition of TGF-β2 enhances the expression of Bcl-2 while reducing the expression of Bax and cleaved caspase 3. This modulation, in turn, helps maintain a balance between pro- and anti-apoptotic signals, reducing excessive cell death and preserving hair follicle function [79].
Mitochondrial regulation of PI3K/AKT signaling
Studies have shown that the activation of the PI3K/AKT pathway enhances mitochondrial activity and promotes cell survival. One of the key outcomes of PI3K/AKT activation is the stimulation of cAMP response element-binding protein (CREB), which directly binds to mitochondrial DNA and facilitates the expression of mitochondrial genes involved in cell survival (such as Bcl-2) and mitochondrial biogenesis (such as PGC-1α) [75]. (For the mitochondrial regulation of hair follicle signaling pathways, see Fig. 2 and Table 1.)
Effects of mitochondrial damage on hair follicle health
Differentiation and barrier maintenance of keratinocytes
Mitochondria play pivotal roles in cellular energy production, ROS generation, and apoptosis, all of which are critical for hair follicle development and maintenance [60]. Studies have demonstrated that mitochondrial dysfunction, particularly in the electron transport chain, can impair hair follicle health by disrupting the delicate balance of cellular metabolism and the stress response [77, 80].
Mitochondrial dysfunction primarily impacts hair follicle cells, such as keratinocytes and DPSCs, by altering their energy metabolism. In particular, keratinocytes require mitochondrial ATP production for their proliferation and differentiation. Impaired mitochondrial function results in decreased ATP levels and increased ROS, which can cause oxidative damage to cellular components, ultimately triggering premature follicle aging or entry into the catagen phase. Moreover, excessive ROS can activate apoptotic pathways, further exacerbating hair follicle cell death and contributing to hair loss [60, 81].
In mouse models with disrupted mitochondrial function, such as those deficient in mitochondrial TFAM, researchers observed significant defects in hair follicle development [77]. These defects are often associated with reduced mitochondrial ROS generation, which is critical for activating signaling pathways, such as the Wnt-β-catenin pathway, which is essential for follicle development and maintenance [82,83,84]. In the absence of functional mitochondria, key signaling molecules fail to activate, resulting in impaired follicle growth and increased susceptibility to external stressors [85].
In addition to ROS, calcium signaling between mitochondria and the endoplasmic reticulum (ER) is vital for maintaining cellular homeostasis [86]. Disruption of mitochondrial calcium uptake can hinder ATP production and activate the mitochondrial permeability transition pore, promoting cell death and further compromising hair follicle health [60].
Hair follicle morphogenesis
CRIF1, a nuclear protein that regulates transcription factors, also plays a crucial role in mitochondrial protein synthesis and the integration of oxidative phosphorylation polypeptides into the mitochondrial membrane [87]. Previous studies have shown that mouse embryonic fibroblasts lacking CRIF1 expression exhibit notable mitochondrial dysfunction, which impacts the transcriptional activation of β-catenin. This dysfunction has been linked to abnormalities in skin differentiation and hair morphology, ultimately delaying the hair growth cycle [88, 89].
AIF is a mitochondrial protein crucial for both energy production and the regulation of apoptosis. In hair follicles, AIF.
facilitates the transition from the anagen phase to the catagen phase by initiating apoptosis upon its release from the mitochondria. Mitochondrial dysfunction, which impairs normal AIF activity, can result in irregular hair follicle cycles, ultimately contributing to hair loss or abnormal growth [90].
In mitochondrial disorders such as Friedreich ataxia (FRDA), frataxin deficiency leads to oxidative stress and compromised hair follicle structure, resulting in thin, weak hair shafts and cuticular damage [91]. ROS generated by dysfunctional mitochondria disrupt keratinocyte differentiation and the hair growth cycle, contributing to hair loss. These findings suggest that mitochondrial dysfunction directly causes oxidative damage, leading to structural abnormalities in hair follicles and subsequent hair loss [92, 93].
Krtap11-1 is a protein located in mitochondria that plays a key role in hair follicle development by regulating signaling pathways such as the Wnt and BMP pathways. Its expression varies during different phases of anagen, peaking during the catagen and telogen phases. Mitochondrial dysfunction can impair the normal function of Krtap11-1, leading to issues in keratinocyte differentiation and oxidative stress regulation. This can result in weak hair shafts and hair loss, highlighting the importance of proper mitochondrial function for hair follicle health [64].
The MPZL3 protein is a mitochondrially localized, nuclear-encoded protein that plays a pivotal role in regulating key physiological processes, including ROS production, lipid metabolism, and energy balance [94]. Disruption of MPZL3 function, as observed in MPZL3 global knockout (GKO) mice, results in premature entry into the anagen phase and various skin abnormalities. The ability of MPZL3 to regulate mitochondrial activity is essential for maintaining the balance between cellular differentiation and homeostasis, particularly in the sebaceous glands of hair follicles. Dysfunctional MPZL3 impairs mitochondrial dynamics, disrupts hair follicle cycling and contributes to conditions such as seborrheic dermatitis-like skin inflammation [8].
Cutaneous energy metabolism and its stressors
HFs operate through an internal metabolic cycle akin to the Cori cycle, which plays a key role in hair growth and development. This study revealed that human hair follicles store glycogen, particularly in the ORS, and that this glycogen is metabolized to generate energy essential for the maintenance of hair follicles. The presence and breakdown of glycogen in the ORS is critical during the hair cycle, with changes observed during different stages, such as anagen and catagen. Mitochondrial dysfunction affecting glycogen metabolism could disrupt energy production, thereby impacting hair growth and leading to abnormalities such as premature onset of catagen [95].
Mitochondria support both anaerobic and aerobic respiration. HFSCs predominantly rely on anaerobic mitochondrial metabolism [96], whereas differentiated cells favour aerobic metabolism. Aerobic respiration in mitochondria is essential for the differentiation and functionality of HFSCs. As HFSCs undergo differentiation, there is a transition in mitochondrial activity from glycolysis to oxidative phosphorylation to meet the increased energy demands. Mitochondrial dysfunction, particularly impaired oxidative phosphorylation, can delay hair regeneration and negatively impact hair follicle health [97,98,99,100]. In addition to respiration, mitochondria significantly contribute to other energy processes, thereby enhancing the energy supply derived from glycolysis, which serves as the primary energy source for hair follicles [96].
Inhibiting the entry of pyruvate into mitochondria disrupts normal oxidative metabolism and accelerates the hair growth cycle, stimulating hair follicle regrowth even in conditions of alopecia [101, 102]. Furthermore, enzymes involved in fatty acid synthesis and elongation, such as stearoyl-CoA desaturase (SCD), are crucial for lipid production in sebaceous glands and skin, which support hair follicle hydration and maintain barrier integrity. Deficiencies in fatty acid metabolism can impair the hair growth cycle, lead to structural damage, and reduce the viability of hair follicles [103, 104].
Novel neuroendocrine regulation in hair follicles
Research interest in the impact of neurohormonal signaling on skin physiology has grown, particularly regarding how hair follicles function as endocrine organs influenced by the hypothalamus‒pituitary‒thyroid (HPT) axis [11]. Thyroid hormones play a vital role in regulating hair follicle physiology by prolonging the anagen phase, promoting keratinocyte proliferation, and affecting pigmentation [10, 105]. Notably, mitochondria not only serve as cellular "powerhouses" but also perform endocrine functions, providing cells with the necessary energy and signals for their function [106]. Thyroid hormones enhance mitochondrial function, improving hair follicle growth, thermogenesis, keratinocyte proliferation, and keratin expression [82, 107, 108]. Previous research has demonstrated increased expression of mitochondria-encoded cytochrome c oxidase subunit 1 (MTCO1) in human scalp hair follicles cultured under the influence of the HPT axis [109]. Thyroid hormones and associated neuroendocrine signals stimulate mitochondrial biogenesis and activity, particularly in the ORS of hair follicles. Mitochondrial dysfunction, resulting from imbalances in hormonal control, can impair ATP production, leading to reduced energy availability for hair follicle maintenance and growth. This disruption may negatively affect the hair growth cycle, potentially contributing to hair loss due to insufficient mitochondrial energy output [108]. (The effects of mitochondrial damage on hair follicle health are illustrated in Fig. 3.)
Involvement of mitochondrial dysfunction in hair-related disorders
Mitochondrial dysfunction is recognized as a significant contributor to various hair diseases. In conditions such as lichen planopilaris, notable mitochondrial abnormalities have been observed in external hair follicle stem cells (EHFSCs) [110, 111]. These abnormalities are consistent with skin-related manifestations, including hair and pigmentation issues, rashes, and acrocyanosis, which occur in approximately 10% of individuals with primary mitochondrial diseases [74].
Mitochondrial homeostasis is intricately regulated by Sirtuin 1 (SIRT1), a histone deacetylase that is primarily activated by the MAPK/ERK/Mfn2 pathway. Reduced expression of SIRT1 results in mitochondrial dysfunction, which triggers an inflammatory stress response mediated by TNF-α in HFSCs. This dysfunctional cascade is particularly pronounced in conditions such as AGA, where DHT induces mitochondrial dysfunction, ultimately leading to aging of the DPCs in AGA [112].
Research has revealed that children with mitochondrial diseases frequently display symptoms such as slow hair growth and fragile, thinning hair [6]. This is likely due to mitochondrial dysfunction, which impairs oxidative phosphorylation and results in a decreased energy supply to hair follicle cells. This disruption affects normal hair follicle cycling and regeneration. One study reported that individuals with progressive mitochondrial encephalopathy exhibit fragile hair characterized by longitudinal grooves and a loss of keratinocytes [113]. Additionally, research on FRDA has underscored its effects on hair health, as both carriers and patients frequently present with fragile hair follicles [91]. These findings emphasize the significant role of mitochondrial dysfunction in various hair disorders.
Mitochondrial oxidative phosphorylation is crucial for ATP generation and the regulation of ROS levels to support the differentiation of keratinocytes. Both aerobic and anaerobic respiration within mitochondria contribute to the proliferation and activity of hair follicle cells. Mitochondrial TFAM influences the transcription of the β-catenin signaling pathway, whereas the mitochondrial ribosomal protein CRIF1 impacts both β-catenin signaling and the hair follicle growth cycle. AIF, located in the mitochondrial inner membrane, triggers caspase-independent apoptosis in follicle cells. The hydrophilic protein Krtap11-1 in mitochondria significantly influences the WNT and BMP signaling pathways. The myelin protein zero-like 3 (MPZL3), a nuclear-encoded mitochondrial protein, also affects the hair growth cycle. Additionally, mitochondrial-encoded cytochrome c oxidase subunit 1 (MTCO1) expression is upregulated by the HPT axis within hair follicles, thereby promoting their growth and development.
Treatment progress
Multiple studies have demonstrated the effectiveness of specific medications in managing hair loss associated with mitochondrial dysfunction, which has sparked a variety of new research objectives in this field.
Hair follicle-targeted therapies
DHT plays a role in accelerating the aging process in DPCs by inducing mitochondrial dysfunction, leading to hair loss [114, 115]. Therapies such as stem cell treatment and hair bioengineering, particularly the use of micrografts containing autologous hair follicle mesenchymal stem cells (HF-MSCs), provide safe and effective methods for improving hair count and density in AGA patients [21]. Platelet-rich plasma (PRP) therapy, which harnesses the growth factors present in platelets, has been shown to stimulate HFSCs, promote new hair follicles and significantly improve hair count, density, and scalp thickness in individuals with AGA [2].
Various drugs target specific signaling pathways in hair follicles to promote hair growth. For example, LSESR activates TGF-β/Smad signaling and reduces DHT-induced inflammation in areas of hair loss [79]. Quercitrin (quercetin-3-O-rhamnoside) activates MAPK/CREB signaling, enhancing cellular energy metabolism and growth factor secretion [75]. The monoterpenoid loliolide stimulates hair growth by activating the AKT and Wnt/β-catenin signaling pathways in DPCs [22]. The active compound of the rice capsule (ACPI) increases Wnt/β-catenin signaling, promoting hair growth and prolonging the anagen phase of the hair cycle. Cosmetic formulations such as ShPI and HtPI containing ACPI can prevent hair aging, reduce COL17A1 degradation in hair follicles exposed to ultraviolet radiation, and regulate androgen metabolism by decreasing DHT production [28]. Additionally, lithium chloride has been found to ameliorate follicular defects related to β-catenin transcriptional issues, resulting in increased follicular length [77].
Mitochondrion–targeted therapies
Mitochondrial function is regulated by a variety of hormones and plays a crucial role in maintaining skin and cellular health, especially in treatment and stress situations. For example, thyroid hormones stimulate mitochondrial biogenesis [107], glucocorticoids increase mitochondrial function to support cell energy requirements during acute stress [116], and vitamin D3 derivatives impact tissue differentiation and the function of various tissues [117], including skin health, by controlling the calcium balance. However, caution is essential when these treatments are used because of their potential toxic effects on mitochondria [118].
Cyanidin-3-O-arabinoside (C3A) has been identified as a natural compound that mitigates the slowing of hair growth induced by DHT and enhances the proliferation of HFSCs by preventing the accumulation of mitochondrial calcium [115]. Minoxidil may address AGA by elevating intracellular calcium levels and increasing ATP synthase activity, thereby fostering hair growth and preventing follicular atrophy [119]. Fisetin activates the SIRT1-mitochondrial axis and enhances keratinocyte activity [120], promoting hair growth [121, 122]. Melatonin, known for its recognized antioxidant and DNA repair properties [61, 123], effectively addresses mitochondrial dysfunction, diminishes ROS production, and minimizes hair follicle damage by assisting in the deacetylation of P53 proteins through SIRT1 to counteract DNA damage [124, 125].
Recent advances in low-intensity laser therapy have shown promising results in promoting hair follicle development and regeneration. By utilizing the ability of mitochondria to absorb photons to increase ATP production and nitric oxide release, this therapy improves blood circulation and increases ROS levels, creating an optimal environment for hair growth [126]. Specific laser settings effectively stimulate DPC proliferation and upregulate the expression of important hair development genes, such as Sox2 and Fgf7 [127,128,129].
Moreover, novel drug delivery methods are attracting increasing interest. Microneedle technology has shown promising therapeutic effects and has been applied in the treatment of AGA [130]. A recent study introduced a microneedle patch integrated with cerium nanozymes (CeNZ), which reshaped the microenvironment around hair follicles by reducing oxidative stress and promoting angiogenesis. This innovative delivery method not only enables precise delivery of CeNZs but also stimulates vascularization around hair follicles, supporting hair regrowth more effectively than traditional treatments such as minoxidil [131,132,133,134].
PPARγ signaling is essential for regulating human hair follicle growth. MHY553, a PPARγ agonist [135], has garnered increased attention in dermatological research because of its ability to activate PPARγ in the skin, leading to the elimination of ROS and decreased expression of inflammatory cytokines [121].
ROS scavengers have been shown to be effective in treating skin conditions associated with high ROS levels [136]. Several clinical trials have investigated the use of antioxidants for the treatment of skin diseases [74]. Targeted antioxidants, such as mitoquinone (MitoQ), demonstrate promising therapeutic effects by efficiently entering mitochondria, which is enhanced by the inherent accumulation of iron in mitochondria [136].
Stem cell regeneration therapy has potential for treating conditions such as hair loss, and adipose-derived stem cell (ADSC) therapy has been shown to stimulate hair growth through the promotion of mitochondrial division [137]. However, elucidating the molecular pathways within the inflammatory microenvironment that can deplete stem cells to improve the efficacy of stem cell therapy is essential [78].(The advances in the treatment of alopecia are mediated by mitochondrial dysfunction are illustrated in Table 2.)
Perspectives
Research on mitochondrial dysfunction in AGA is rapidly advancing, offering new therapeutic targets that focus on restoring mitochondrial health within hair follicles. These developments pave the way for innovative treatments aimed at enhancing mitochondrial function, which is critical for energy production and the survival of hair follicle cells. By focusing on mitochondrial-targeted therapies, researchers can explore new strategies for addressing AGA, shifting the focus from traditional approaches to targeting the underlying cellular dysfunction. These advancements offer promising prospects for more effective interventions in hair loss treatment. (Promising treatment strategies for AGA are illustrated in Fig. 4.)
Potential therapeutic targets in hair follicles
Recent studies have revealed significant findings indicating potential new therapeutic targets in hair follicles. SCUBE3, a nontraditional TGF-β ligand, is essential for hair follicle growth, as it is secreted by DP fibroblasts and promotes hair growth. The expression pattern and function of SCUBE3 are partially conserved in human scalp hair follicles, suggesting that SCUBE3 expression could be a promising target for hair follicle therapy [74]. Additionally, Lepr has been identified as a characteristic gene of DP fibroblasts and is currently being investigated for targeted drug delivery in hair follicles. The LeprB-Cre genetic tool has been shown to be effective in targeting DP fibroblasts, indicating the potential for developing customized drug delivery systems for hair follicles on the basis of the specific expression of Lepr in these cells [138, 139]. Targeting DPC offers new strategies for treating hair loss. Activating Hh signaling promotes fibroblast states crucial for hair growth, including DP-like fibroblasts and adipocyte-fibroblast precursors, thus identifying a novel target for hair follicle therapies [138]. Biomarkers related to hair loss, identified through histologic techniques could serve as key targets for future treatments [140].
Targeting adipocytes for hair regeneration
Studies have shown that whole-layer craniomaxillofacial skin reconstruction using human-derived adipose stem cells and an extracellular matrix led to the development of hair follicle-like structures in regenerated tissues [141]. This discovery highlights the significant role that adipocytes play in hair follicle regeneration. The interaction between hair follicles and dermal white adipose tissue (dWAT) is crucial for the hair growth cycle [142, 143]. Further research has shown that dWAT secretes hepatocyte growth factor (HGF), which promotes hair follicle growth and pigmentation by activating the Wnt/β-catenin signaling pathway in hair matrix keratinocytes [144]. Additionally, dermal adipocytes are believed to shift from lipogenesis during the anagen phase to lipophagy and lipolysis during the catagen phase, thereby releasing free glycerol and cholesterol. This metabolic switch plays a regulatory role in the hair growth cycle [145]. Moreover, adipose stem cell extracts have demonstrated effectiveness in treating AGA [146], further emphasizing the importance of targeting adipocytes and their metabolic processes as a promising approach for AGA treatment.
Targeting mitochondrial receptors
Research has expanded to include mitochondrial targets for treating hair loss, as mitochondrial oxidative stress plays a significant role in conditions such as AGA. Key targets, such as SIRT1 and MPZL3, which are crucial for metabolism and stress responses, show promise in addressing hair follicle dysfunction [121, 135]. The Sirtuin family of proteins, particularly Sirtuins 1 and 3, has been recognized for their ability to reduce oxidative stress and influence keratinocyte differentiation. However, knowledge of specific activators of Sirt3 is lacking, and Sirt1 is the primary means of activation [147]. MPZL3, a protein localized to mitochondria, has been identified as an important regulator of the hair follicle cycle and plays a role in hair growth regulation as a downstream effect of PPARγ. PPARγ is a promising target for the treatment of mitochondrial dermatoses, and its use as a downstream effector of PPARγ could enhance the specificity of dermatological treatment and minimize adverse effects [74]. Recent research has suggested that cannabinoid receptor type 1 (CB1) inhibits mitochondrial function in keratinocyte-forming cells [148], with its activation in cell and mitochondrial membranes impacting hair growth [74]. This has led to speculation about the potential benefits of local therapeutic interventions through the selective activation of mtCB1 to reduce excessive mitochondrial ROS production resulting from dysregulated mitochondrial activity, thereby promoting hair growth.
Mitochondrial antioxidants
In addition, mitochondrial antioxidants such as MitoQ and SkQ1 offer potential solutions for the treatment of AGA. MitoQ, known for its antioxidant properties, has demonstrated therapeutic effects in neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease, whereas SkQ1, like MitoQ, is used in the cosmetic industry for its antiaging properties and ability to scavenge ROS and enhance cell viability. Mito-TEMPO and Mito-VitE are both powerful antioxidants with distinct mechanisms of action. Mito-TEMPO reduces ROS levels to prevent ROS-induced damage, while Mito-VitE effectively protects cells from oxidative stress and apoptosis [149]. A recent study demonstrated that N-acetylcysteine (NAC), which targets mitochondrial oxidative stress through its ROS-scavenging properties, effectively improved hair growth in men with early-onset AGA. The ability of NAC to reduce mitochondrial oxidative damage suggests its potential as a mitochondrial-targeted therapy for AGA when used alone or combined with minoxidil [150].
In addition to the role of antioxidants, recent advancements in mitochondrial drug delivery systems have markedly improved therapeutic efficacy. Conventional approaches, such as the use of triphenylphosphine (TPP), target mitochondria by interacting with the negatively charged mitochondrial membrane, thereby facilitating the delivery of lipophilic antioxidants such as MitoQ and Mito-TEMPO [149, 151,152,153,154]. Szeto-Schiller (SS) peptides, which accumulate on the inner mitochondrial membrane through electrostatic and hydrophobic interactions, exhibit lower toxicity and have demonstrated therapeutic benefits across various experimental models [151, 153]. Furthermore, nanocarrier-based systems, including MITO-Porter liposomes and self-assembled cationic carriers such as DQAsomes, broaden the potential for mitochondrial targeting by allowing precise antioxidant delivery [151,152,153,154,155,156]. Additionally, researchers have developed a fluorinated amphiphilic core–shell micelle system that enhances mitochondrial targeting by binding to mitochondrial phospholipids, particularly cardiolipin, and operates independently of membrane potential, thereby addressing the limitations associated with traditional cationic ligands [157].
In addition to the use of traditional antioxidants and drug delivery systems, emerging technologies such as microneedle-assisted delivery, have further broadened the possibilities for AGA treatment. Cerium oxide nanozymes have been integrated with microneedles, enabling antioxidants to be delivered approximately 300 microns deep into the skin for rapid drug release [131]. Recent research has improved the stability and targeting efficacy of CeNZ cerium nanoparticles by utilizing systems such as mesoporous silica carriers and graphdiyne-based nanocomposites [132, 133]. The integration of conventional antioxidants with advanced drug delivery systems, alongside novel methodologies such as microneedles, has established a robust framework for the development of mitochondrion-targeting therapies for AGA. Clinical studies have underscored the benefits of antioxidants in hair treatment. For example, a 24-week randomized, double-blind, placebo-controlled trial demonstrated that a topical regimen containing functional antioxidants significantly improved scalp condition, reduced hair shedding, and increased hair retention [158, 159]. However, while reducing oxidative stress is beneficial, it is essential to recognize that ROS also play a regulatory role in hair follicle cycling. Excessive suppression of ROS can disrupt these natural signaling pathways. Therefore, the therapeutic rationale for using antioxidants in AGA treatment should emphasize achieving a balance that mitigates oxidative damage without completely eliminating ROS, thus preserving the normal physiological processes of hair follicle growth.
Mitochondrial transfer
Recent research has demonstrated successful mitochondrial transfer in treating mitochondrial dysfunction-related diseases. In one study, researchers were able to transfer functional mitochondria from healthy donor cells to damaged cells, restoring their bioenergetic capacity and reducing oxidative stress [160]. Another study showed that mitochondrial transfer improved cell survival and recovery in models of ischemic injury, helping restore normal cellular metabolism [160].
Given that mitochondrial dysfunction is a critical factor in AGA progression, these findings suggest that mitochondrial transfer could be explored as a novel therapeutic approach for AGA. By replenishing damaged mitochondria in hair follicle cells, mitochondrial transfer may restore energy production, reduce oxidative damage, and support normal hair follicle cycling, offering a new direction for AGA treatment.
The pathogenesis of diffuse androgenetic alopecia, also known as female pattern hair loss, remains unclear [161]. However, studies have indicated an increase in apoptosis within the hair follicles of patients with FPHL [162]. In miniaturized hair follicles, there is elevated nuclear expression of the aryl hydrocarbon receptor [161], which regulates genes involved in mitochondrial biogenesis and function [163]. This regulation promotes keratinocyte apoptosis and contributes to follicle miniaturization. These findings suggest that targeting mitochondrial dysfunction may represent a promising therapeutic approach for FPHL, potentially reducing oxidative stress and mitigating hair follicle apoptosis. Further research is necessary to validate the effectiveness of mitochondrial-targeted therapies in female patients and to investigate their specific mechanisms of action. The focus on targeting mitochondria in AGA offers a promising new direction for treatment strategies. Directly addressing mitochondrial dysfunction within hair follicle cells can improve the cellular energy balance, reduce oxidative stress, and promote hair follicle health. As research advances, these mitochondrial-targeted approaches could revolutionize AGA treatment, marking the beginning of a new era in "mitochondrial dermatology" with potential applications for various skin conditions.
This figure illustrates potential therapies for AGA, focusing on mitochondrial-targeted treatments and the use of antioxidants to reduce oxidative stress. Additionally, mitochondrial transfer offers a novel method for restoring mitochondrial function. In hair follicle-targeted therapies, key targets include SCUBE3, Lepr, and the TGF-β signaling pathway, along with the supportive role of dermal adipose tissue in promoting hair growth.
Availability of data and materials
Not applicable
References
Oiwoh SO, Enitan AO, Adegbosin OT, Akinboro AO, Onayemi EO. Androgenetic alopecia: a review. The Niger Postgrad Med J. 2024;31:85–92. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/npmj.npmj_47_24.
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 Therapy. 2021;12:453. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-021-02527-y.
Kasahara A, Scorrano L. Mitochondria: from cell death executioners to regulators of cell differentiation. Trends in Cell Biol. 2014;24(12):761–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2014.08.005.
Robicsek O, Karry R, Petit I, Salman-Kesner N, Müller F-J, Klein E, Aberdam D, Ben-Shachar D. Abnormal neuronal differentiation and mitochondrial dysfunction in hair follicle-derived induced pluripotent stem cells of schizophrenia patients. Mol Psychiatry. 2013;18(10):1067–76. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/mp.2013.67.
Feichtinger RG, Sperl W, Bauer JW, Kofler B. Mitochondrial dysfunction: a neglected component of skin diseases. Exp Dermatol. 2014;23(9):607–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.12484.
Silengo M, Valenzise M, Spada M, Ferrero GB, Ferraris S, Dassi P, Jarre L. Hair anomalies as a sign of mitochondrial disease. Eur J Pediatrics. 2003;162(7–8):459–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00431-003-1228-5.
Chew EGY, Lim TC, Leong MF, Liu X, Sia YY, Leong ST, et al. Observations that suggest a contribution of altered dermal papilla mitochondrial function to androgenetic alopecia. Exp Dermatol. 2022;31:906–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.14536.
Nicu C, Wikramanayake TC, Paus R. Clues that mitochondria are involved in the hair cycle clock: MPZL3 regulates entry into and progression of murine hair follicle cycling. Exp Dermatol. 2020;29(12):1243–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.14213.
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;15:1647–70. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms15011647.
Oláh A, Gherardini J, Bertolini M, Chéret J, Ponce L, Kloepper J, Bíró T, Soeberdt M, Abels C, Paus R. the thyroid hormone analogue KB2115 (Eprotirome) prolongs human hair growth (Anagen) Ex Vivo. J Invest Dermatol. 2016;136(8):1711–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2016.03.033.
Paus R, Langan EA, Vidali S, Ramot Y, Andersen B. Neuroendocrinology of the hair follicle: principles and clinical perspectives. Trends in Mol Med. 2014;20:559–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molmed.2014.06.002.
Sohn KM, Jeong KH, Kim JE, Park YM, Kang H. Hair growth-promotion effects of different alternating current parameter settings are mediated by the activation of Wnt/β-catenin and MAPK pathway. Exp Dermatol. 2015;24:958–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.12827.
Paus R, Foitzik K. In search of the “hair cycle clock”: a guided tour. Diff; Res Biol Divers. 2004;72:489–511. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1432-0436.2004.07209004.x.
Wang X, Liu Y, He J, Wang J, Chen X, Yang R. Regulation of signaling pathways in hair follicle stem cells. Burns & trauma. 2022;10:22. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/burnst/tkac022.
Li H, Ziemer M, Stojanovic I, Saksida T, Maksimovic-Ivanic D, Mijatovic S, et al. Mesenchymal stem cells from mouse hair follicles reduce hypertrophic scarring in a murine wound healing model. Stem Cell Rev And Reports. 2022;18:2028–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12015-021-10288-7.
Lee JH, Choi S. Deciphering the molecular mechanisms of stem cell dynamics in hair follicle regeneration. Exp Mol Med. 2024;56:110–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-023-01151-5.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-020-0624-8.
Lin Y, Liu C, Zhan X, Wang B, Li K, Li J. Jagged1 and epidermal growth factor promoted androgen-suppressed mouse hair growth in vitro and in vivo. Front Pharmacol. 2019;10:1634. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2019.01634.
Schneider MR, Schmidt-Ullrich R, Paus R. The hair follicle as a dynamic miniorgan. Current Biol CB. 2009;19:132–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cub.2008.12.005.
David R. Development: a good hair day for ROS. Nature Rev Mol Cell Biol. 2013;14:194–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm3547.
Gentile P, Garcovich S. Advances in regenerative stem cell therapy in androgenic alopecia and hair loss: wnt pathway, growth-factor, and mesenchymal stem cell signaling impact analysis on cell growth and hair follicle development. Cells. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells8050466.
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/beta-catenin signaling pathway. J Microbiol Biotechnol. 2019;29:1830–40. https://doiorg.publicaciones.saludcastillayleon.es/10.4014/jmb.1908.08018.
Wier EM, Garza LA. Through the lens of hair follicle neogenesis, a new focus on mechanisms of skin regeneration after wounding. Seminars in cell & Develop Biol. 2020;100:122–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2019.10.002.
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;81:2323–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/09168451.2017.1385383.
Xue Y, Lin L, Li Q, Liu K, Hu M, Ye J, et al. SCD1 Sustains Homeostasis of Bulge Niche via Maintaining Hemidesmosomes in Basal Keratinocytes. Adv Sci (Weinheim, Baden-Wurttemberg, Germany). 2023;10:e2201949. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/advs.202201949.
Jiang Fu, Hsu W. Epidermal Wnt controls hair follicle induction by orchestrating dynamic signaling crosstalk between the epidermis and dermis. J Invest Dermatol. 2013;133(4):890–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2012.407.
McNeill H, Woodgett JR. When pathways collide: collaboration and connivance among signalling proteins in development. Nat Rev Mol Cell Biol. 2010;11(6):404–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm2902.
Honda Igarashi M, da Silva SG, Mercuri M, Zuardi F, Facchini G, da Silva GH, et al. Novel complex of cosmetic ingredients with promising action in preventing hair loss and follicular aging through mechanism involving enrichment of WNT/signaling, mitochondrial activity, and stem cells maintenance. J Cosmetic Dermatol. 2021;20:2179–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jocd.13815.
Lei M, Lai X, Bai X, Qiu W, Yang T, Liao X, et al. Prolonged overexpression of Wnt10b induces epidermal keratinocyte transformation through activating EGF pathway. Histochem Cell Biol. 2015;144:209–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00418-015-1330-6.
Li YH, Zhang K, Yang K, Ye JX, Xing YZ, Guo HY, et al. Adenovirus-mediated Wnt10b overexpression induces hair follicle regeneration. The J Investigat Dermatol. 2013;133:42–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2012.235.
Owens P, Han G, Li AG, Wang XJ. The role of Smads in skin development. The J Invest Dermatol. 2008;128:783–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.jid.5700969.
Tzavlaki K, Moustakas A. TGF-β Signaling. Biomolecules. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom10030487.
Hansdah K, Singh N, Bouzid A, Priyadarshi S, Ray CS, Desai A, et al. Evaluation of the genetic association and mRNA expression of the COL1A1, BMP2, and BMP4 genes in the development of otosclerosis. Gen Testing And Mol Biomark. 2020;24:343–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/gtmb.2019.0235.
Plikus MV, Mayer JA, de la Cruz D, Baker RE, Maini PK, Maxson R, et al. Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature. 2008;451:340–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature06457.
Xiao L, Yuan X, Sharkis SJ. Activin A maintains self-renewal and regulates fibroblast growth factor, Wnt, and bone morphogenic protein pathways in human embryonic stem cells. Stem Cells. 2006;24(6):1476–86. https://doiorg.publicaciones.saludcastillayleon.es/10.1634/stemcells.2005-0299.
Huelsken J, Vogel R, Erdmann B, Cotsarelis G, Birchmeier W. beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105:533–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0092-8674(01)00336-1.
Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors. Cell. 2004;116(6):769–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0092-8674(04)00255-7.
Ge M, Liu C, Li L, Lan M, Yingying Y, Lijie G, Yang S, Zhang K, Zhang Y, Wang T, Liu C, Liu F, Li M, Xiong L, Wang K, He T, Dai Y, Zhao Y, Li N, Zhengquan Y, Meng Q. miR-29a/b1 inhibits hair follicle stem cell lineage progression by spatiotemporally suppressing WNT and BMP signaling. Cell Rep. 2019;29(8):2489-2504.e4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2019.10.062.
Aubin-Houzelstein G. Notch signaling and the developing hair follicle. In: Reichrath J, Reichrath S, editors. Notch Signaling in Embryology and Cancer. New York: Springer US; 2012. p. 142–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-1-4614-0899-4_11.
Shi Y, Shu B, Yang R, Xu Y, Xing B, Liu J, et al. Wnt and Notch signaling pathway involved in wound healing by targeting c-Myc and Hes1 separately. Stem Cell Res Therapy. 2015;6:120. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-015-0103-4.
Vauclair S, Nicolas M, Barrandon Y, Radtke F. Notch1 is essential for postnatal hair follicle development and homeostasis. Develop Biol. 2005;284(1):184–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ydbio.2005.05.018.
Brownell I, Guevara E, Bai CB, Loomis CA, Joyner AL. Nerve-derived sonic hedgehog defines a niche for hair follicle stem cells capable of becoming epidermal stem cells. Cell Stem Cell. 2011;8:552–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2011.02.021.
Pak E, Segal RA. Hedgehog signal transduction: key players, oncogenic drivers, and cancer therapy. Develop Cell. 2016;38(4):333–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2016.07.026.
Abe Y, Tanaka N. Roles of the hedgehog signaling pathway in epidermal and hair follicle development, homeostasis, and cancer. J Develop Biol. 2017;5(4):12. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jdb5040012.
Sun X, Are A, Annusver K, Sivan U, Jacob T, Dalessandri T, Joost S, Füllgrabe A, Gerling M, Kasper M. Coordinated hedgehog signaling induces new hair follicles in adult skin. Elife. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.46756.
Peterson SC, Eberl M, Vagnozzi AN, Belkadi A, Veniaminova NA, Verhaegen ME, et al. Basal cell carcinoma preferentially arises from stem cells within hair follicle and mechanosensory niches. Cell Stem Cell. 2015;16:400–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.stem.2015.02.006.
Qian H, Cao P, Hu M, Gao S, Yan N, Gong X. Inhibition of tetrameric patched1 by sonic hedgehog through an asymmetric paradigm. Nat Commun. 2019;10:2320. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-019-10234-9.
Hers I, Vincent EE, Tavaré JM. Akt signalling in health and disease. Cell signal. 2011;23:1515–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellsig.2011.05.004.
Chen Y, Fan Z, Wang X, Mo M, Zeng SB, Xu RH, et al. PI3K/Akt signaling pathway is essential for de novo hair follicle regeneration. Stem cell Rese Therapy. 2020;11:144. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-020-01650-6.
Hardy KM, Yatskievych TA, Konieczka J, Bobbs AS, Antin PB. FGF signalling through RAS/MAPK and PI3K pathways regulates cell movement and gene expression in the chicken primitive streak without affecting E-cadherin expression. BMC Develop Biol. 2011;11:20. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-213x-11-20.
Koyasu S. The role of PI3K in immune cells. Nat Immunol. 2003;4:313–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni0403-313.
Bai T, Liu F, Zou F, Zhao G, Jiang Y, Liu L, et al. Epidermal growth factor induces proliferation of hair follicle-derived mesenchymal stem cells through epidermal growth factor receptor-mediated activation of ERK and AKT signaling pathways associated with upregulation of cyclin D1 and downregulation of p16. Stem Cells And Develop. 2017;26:113–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/scd.2016.0234.
Cai B, Zheng Y, Ma S, Xing Q, Wang X, Yang B, et al. Long non-coding RNA regulates hair follicle stem cell proliferation and differentiation through PI3K/AKT signal pathway. Mol Med Reports. 2018;17:5477–83. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/mmr.2018.8546.
Akilli Ozturk O, Pakula H, Chmielowiec J, Qi J, Stein S, Lan L, et al. Gab1 and mapk signaling are essential in the hair cycle and hair follicle stem cell quiescence. Cell Rep. 2015;13:561–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2015.09.015.
Sharma P, Sampath H. Mitochondrial DNA Integrity: Role in Health and Disease. Cells. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells8020100.
Luft R, Landau BR. Mitochondrial medicine. J Int Med. 1995;238:405–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2796.1995.tb01218.x.
De Stefani D, Bononi A, Romagnoli A, Messina A, De Pinto V, Pinton P, et al. VDAC1 selectively transfers apoptotic Ca2+ signals to mitochondria. Cell Death Diff. 2012;19:267–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cdd.2011.92.
Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell. 2007;131:596–610. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2007.08.036.
Csordás G, Weaver D, Hajnóczky G. Endoplasmic reticulum-mitochondrial contactology: structure and signaling functions. Trends in Cell Biol. 2018;28:523–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2018.02.009.
Csordás G, Hajnóczky G. SR/ER-mitochondrial local communication: calcium and ROS. Biochimica et Biophys Acta. 2009;1787:1352–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbabio.2009.06.004.
Galano A, Tan DX, Reiter RJ. Melatonin: a versatile protector against oxidative DNA damage. Molecules (Basel, Switzerland). 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules23030530.
Booth DM, Enyedi B, Geiszt M, Várnai P, Hajnóczky G. Redox nanodomains are induced by and control calcium signaling at the ER-mitochondrial interface. Mol Cell. 2016;63:240–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.molcel.2016.05.040.
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. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-022-00800-7.
Liang S, Bao Z, Zhao B, Zhou T, Li J, Liu M, et al. Characterization and functional analysis of Krtap11–1 during hair follicle development in Angora rabbits (Oryctolagus cuniculus). Gen Genom. 2020;42:1281–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13258-020-00995-9.
Hussein MR, Abu-Dief EE, Abd El-Reheem MH, Abd-Elrahman A. Ultrastructural evaluation of the radioprotective effects of melatonin against X-ray-induced skin damage in Albino rats. Int J Exp Pathol. 2005;86:45–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.0959-9673.2005.00412.x.
Lu Z, Fischer TW, Hasse S, Sugawara K, Kamenisch Y, Krengel S, et al. Profiling the response of human hair follicles to ultraviolet radiation. The J Invest Dermatol. 2009;129:1790–804. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2008.418.
Hussein MR, Haemel AK, Wood GS. Apoptosis and melanoma: molecular mechanisms. The J Pathol. 2003;199:275–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.1300.
Hussein MR, Haemel AK, Wood GS. p53-related pathways and the molecular pathogenesis of melanoma. Eur J Cancer Prevent : The Official J Eur Cancer Prevent Org (ECP). 2003;12:93–100. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/00008469-200304000-00002.
Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature. 1999;397:441–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/17135.
Gao S, Hu J. Mitochondrial fusion: the machineries in and out. Trends in Cell Biol. 2021;31:62–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tcb.2020.09.008.
Tilokani L, Nagashima S, Paupe V, Prudent J. Mitochondrial dynamics: overview of molecular mechanisms. Essays in Biochem. 2018;62:341–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/ebc20170104.
Serasinghe MN, Chipuk JE. Mitochondrial fission in human diseases. Handbook Exp Pharmacol. 2017;240:159–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/164_2016_38.
Onishi M, Yamano K, Sato M, Matsuda N, Okamoto K. Molecular mechanisms and physiological functions of mitophagy. The EMBO J. 2021;40:e104705. https://doiorg.publicaciones.saludcastillayleon.es/10.15252/embj.2020104705.
Wikramanayake TC, Chéret J, Sevilla A, Birch-Machin M, Paus R. Targeting mitochondria in dermatological therapy: beyond oxidative damage and skin aging. Expert Opinion On Therapeut Targets. 2022;26:233–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/14728222.2022.2049756.
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 (Basel, Switzerland). 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules25174004.
Mitra K, Rikhy R, Lilly M, Lippincott-Schwartz J. DRP1-dependent mitochondrial fission initiates follicle cell differentiation during Drosophila oogenesis. The J Cell Biol. 2012;197:487–97. https://doiorg.publicaciones.saludcastillayleon.es/10.1083/jcb.201110058.
Hamanaka RB, Glasauer A, Hoover P, Yang S, Blatt H, Mullen AR, et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci signal. 2013;6:8. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scisignal.2003638.
Liu J, Xu Y, Wu Q, Ding Q, Fan W. Sirtuin-1 protects hair follicle stem cells from TNFα-mediated inflammatory stress via activating the MAPK-ERK-Mfn2 pathway. Life Sci. 2018;212:213–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2018.10.003.
Zhu HL, Gao YH, Yang JQ, Li JB, Gao J. Serenoa repens extracts promote hair regeneration and repair of hair loss mouse models by activating TGF-β and mitochondrial signaling pathway. Eur Review Med Pharmacol Sci. 2018;22:4000–8. https://doiorg.publicaciones.saludcastillayleon.es/10.26355/eurrev_201806_15285.
Baris OR, Klose A, Kloepper JE, Weiland D, Neuhaus JF, Schauen M, et al. The mitochondrial electron transport chain is dispensable for proliferation and differentiation of epidermal progenitor cells. Stem Cells (Dayton, Ohio). 2011;29:1459–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/stem.695.
Yoo JA, Yu E, Park SH, Oh SW, Kwon K, Park SJ, et al. Blue light irradiation induces human keratinocyte cell damage via transient receptor potential vanilloid 1 (TRPV1) regulation. Oxid Med Cell Longev. 2020;2020:8871745. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2020/8871745.
Kloepper JE, Baris OR, Reuter K, Kobayashi K, Weiland D, Vidali S, et al. Mitochondrial function in murine skin epithelium is crucial for hair follicle morphogenesis and epithelial-mesenchymal interactions. The J Invest Dermatol. 2015;135:679–89. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2014.475.
Hamanaka RB, Chandel NS. Mitochondrial metabolism as a regulator of keratinocyte differentiation. Cell Logist. 2013;3:e25456. https://doiorg.publicaciones.saludcastillayleon.es/10.4161/cl.25456.
Purba TS, Brunken L, Peake M, Shahmalak A, Chaves A, Poblet E, et al. Characterisation of cell cycle arrest and terminal differentiation in a maximally proliferative human epithelial tissue: lessons from the human hair follicle matrix. Eur J Cell Biol. 2017;96:632–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejcb.2017.03.011.
Elias P, Ahn S, Brown B, Crumrine D, Feingold KR. Origin of the epidermal calcium gradient: regulation by barrier status and role of active vs passive mechanisms. The J Invest Dermatol. 2002;119:1269–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1523-1747.2002.19622.x.
Lemasters JJ, Ramshesh VK, Lovelace GL, Lim J, Wright GD, Harland D, et al. Compartmentation of mitochondrial and oxidative metabolism in growing hair follicles: a ring of fire. The J Invest Dermatol. 2017;137:1434–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2017.02.983.
Bescos R, Boden MJ, Jackson ML, Trewin AJ, Marin EC, Levinger I, et al. Four days of simulated shift work reduces insulin sensitivity in humans. Acta Physiol (Oxf). 2018;223:e13039. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/apha.13039.
Shin JM, Choi DK, Sohn KC, Kim JY, Im M, Lee Y, et al. Targeted deletion of Crif1 in mouse epidermis impairs skin homeostasis and hair morphogenesis. Sci Rep. 2017;7:44828. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep44828.
Shin JM, Ko JW, Choi CW, Lee Y, Seo YJ, Lee JH, et al. Deficiency of Crif1 in hair follicle stem cells retards hair growth cycle in adult mice. PLoS ONE. 2020;15:e0232206. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0232206.
Lan S, Liu F, Zhao G, Zhou T, Wu C, Kou J, et al. Cyclosporine A increases hair follicle growth by suppressing apoptosis-inducing factor nuclear translocation: a new mechanism. Fund Clin Pharmacol. 2015;29:191–203. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/fcp.12100.
Turkmenoglu FP, Kasirga UB, Celik HH. Ultra-structural hair alterations in Friedreich’s ataxia: a scanning electron microscopic investigation. Microscopy Res Tech. 2015;78:731–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jemt.22531.
Pandolfo M. Friedreich ataxia: the clinical picture. J Neurol. 2009;256(1):3–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00415-009-1002-3.
Cook A, Giunti P. Friedreich’s ataxia: clinical features, pathogenesis and management. British Med Bullet. 2017;124:19–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bmb/ldx034.
Wikramanayake TC, Nicu C, Chéret J, Czyzyk TA, Paus R. Mitochondrially localized MPZL3 emerges as a signaling hub of mammalian physiology. BioEssays : News Rev Mol, Cell Develop Biol. 2021;43:e2100126. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/bies.202100126.
Figlak K, Williams G, Bertolini M, Paus R, Philpott MP. Human hair follicles operate an internal Cori cycle and modulate their growth via glycogen phosphorylase. Sci Rep. 2021;11:20761. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-99652-8.
Randall VA, Sundberg JP, Philpott MP. Animal and in vitro models for the study of hair follicles. The J Invest Dermatol Symp Proceed. 2003;8:39–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1523-1747.2003.12170.x.
Hsu P, Qu CK. Metabolic plasticity and hematopoietic stem cell biology. Current Opinion Hematol. 2013;20:289–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/MOH.0b013e328360ab4d.
Jang H, Yang J, Lee E, Cheong JH. Metabolism in embryonic and cancer stemness. Archives of Pharmacal Res. 2015;38:381–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12272-015-0558-y.
Kondoh H, Lleonart ME, Nakashima Y, Yokode M, Tanaka M, Bernard D, et al. A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid Redox Signal. 2007;9:293–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/ars.2006.1467.
Tang Y, Luo B, Deng Z, Wang B, Liu F, Li J, et al. Mitochondrial aerobic respiration is activated during hair follicle stem cell differentiation, and its dysfunction retards hair regeneration. PeerJ. 2016;4:e1821. https://doiorg.publicaciones.saludcastillayleon.es/10.7717/peerj.1821.
Armstrong L, Tilgner K, Saretzki G, Atkinson SP, Stojkovic M, Moreno R, et al. Human induced pluripotent stem cell lines show stress defense mechanisms and mitochondrial regulation similar to those of human embryonic stem cells. Stem cells (Dayton, Ohio). 2010;28:661–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/stem.307.
Flores A, Choi S, Hsu YC, Lowry WE. Inhibition of pyruvate oxidation as a versatile stimulator of the hair cycle in models of alopecia. Exp Dermatol. 2021;30:448–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.14307.
Feingold KR, Elias PM. Role of lipids in the formation and maintenance of the cutaneous permeability barrier. Biochimica et Biophys Acta. 2014;1841:280–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbalip.2013.11.007.
Wertz PW. Lipids and the permeability and antimicrobial barriers of the skin. J Lipid. 2018;2018:5954034. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2018/5954034.
Messenger AG. Thyroid hormone and hair growth. The Brit J Dermatol. 2000;142:633–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.1365-2133.2000.03521.x.
Picard M, McEwen BS, Epel ES, Sandi C. An energetic view of stress: Focus on mitochondria. Front Neuroendocrinol. 2018;49:72–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.yfrne.2018.01.001.
Vidali S, Chéret J, Giesen M, Haeger S, Alam M, Watson REB, et al. Thyroid hormones enhance mitochondrial function in human epidermis. The J Invest Dermatol. 2016;136:2003–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2016.05.118.
Vidali S, Knuever J, Lerchner J, Giesen M, Bíró T, Klinger M, et al. Hypothalamic-pituitary-thyroid axis hormones stimulate mitochondrial function and biogenesis in human hair follicles. The J Invest Dermatol. 2014;134:33–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2013.286.
Bodó E, Kromminga A, Bíró T, Borbíró I, Gáspár E, Zmijewski MA, et al. Human female hair follicles are a direct, nonclassical target for thyroid-stimulating hormone. The J Invest Dermatol. 2009;129:1126–39. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2008.361.
Hardman-Smart JA, Purba TS, Panicker S, Farjo B, Farjo N, Harries MJ, et al. Does mitochondrial dysfunction of hair follicle epithelial stem cells play a role in the pathobiology of lichen planopilaris? The Brit J Dermatol. 2020;183:964–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bjd.19259.
Harries MJ, Meyer K, Chaudhry I, EK J, Poblet E, Griffiths CE, et al. Lichen planopilaris is characterized by immune privilege collapse of the hair follicle’s epithelial stem cell niche. The J Pathol. 2013;231:236–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/path.4233.
O’Sullivan JDB, Peters EMJ, Amer Y, Atuluru P, Chéret J, Rosenberg AM, et al. The impact of perceived stress on the hair follicle: Towards solving a psychoneuroendocrine and neuroimmunological puzzle. Front Neuroendocrinol. 2022;66:101008. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.yfrne.2022.101008.
Bodemer C, Rötig A, Rustin P, Cormier V, Niaudet P, Saudubray JM, et al. Hair and skin disorders as signs of mitochondrial disease. Pediatrics. 1999;103:428–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1542/peds.103.2.428.
Bahta AW, Farjo N, Farjo B, Philpott MP. Premature senescence of balding dermal papilla cells in vitro is associated with p16(INK4a) expression. The J Invest Dermatol. 2008;128:1088–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.jid.5701147.
Upton JH, Hannen RF, Bahta AW, Farjo N, Farjo B, Philpott MP. Oxidative stress-associated senescence in dermal papilla cells of men with androgenetic alopecia. The J Invest Dermatol. 2015;135:1244–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/jid.2015.28.
Choi GE, Han HJ. Glucocorticoid impairs mitochondrial quality control in neurons. Neurobiol Dis. 2021;152:105301. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2021.105301.
Oda Y, Hu L, Nguyen T, Fong C, Zhang J, Guo P, et al. Vitamin D receptor is required for proliferation, migration, and differentiation of epidermal stem cells and progeny during cutaneous wound repair. The J Invest Dermatol. 2018;138:2423–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2018.04.033.
de Oliveira MR. Vitamin A and retinoids as mitochondrial toxicants. Oxidative Med Cell Longev. 2015;2015:140267. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2015/140267.
Goren A, Naccarato T, Situm M, Kovacevic M, Lotti T, McCoy J. Mechanism of action of minoxidil in the treatment of androgenetic alopecia is likely mediated by mitochondrial adenosine triphosphate synthase-induced stem cell differentiation. J Biol Regul Homeost Agents. 2017;31:1049–53.
Ogawa M, Udono M, Teruya K, Uehara N, Katakura Y. Exosomes derived from fisetin-treated keratinocytes mediate hair growth promotion. Nutrients. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu13062087.
Ramot Y, Alam M, Oláh A, Bíró T, Ponce L, Chéret J, et al. Peroxisome proliferator-activated receptor-γ-mediated signaling regulates mitochondrial energy metabolism in human hair follicle epithelium. The J Invest Dermatol. 2018;138:1656–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2018.01.033.
Sreedhar A, Aguilera-Aguirre L, Singh KK. Mitochondria in skin health, aging, and disease. Cell Death Dis. 2020;11:444. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-020-2649-z.
Hendrix S, Handjiski B, Peters EM, Paus R. A guide to assessing damage response pathways of the hair follicle: lessons from cyclophosphamide-induced alopecia in mice. The J Invest Dermatol. 2005;125:42–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.0022-202X.2005.23787.x.
He R, Cui M, Lin H, Zhao L, Wang J, Chen S, et al. Melatonin resists oxidative stress-induced apoptosis in nucleus pulposus cells. Life Sci. 2018;199:122–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2018.03.020.
Xu D, Liu L, Zhao Y, Yang L, Cheng J, Hua R, et al. Melatonin protects mouse testes from palmitic acid-induced lipotoxicity by attenuating oxidative stress and DNA damage in a SIRT1-dependent manner. J Pineal Res. 2020;69:e12690. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jpi.12690.
Jampa-Ngern S, Viravaidya-Pasuwat K, Suvanasuthi S, Khantachawana A. Effect of laser diode light irradiation on growth capability of human hair follicle dermal papilla cells. In: Annual International Conference of the IEEE Engineering in Medicine and Biology Society IEEE Engineering in Medicine and Biology Society Annual International Conference. 2017;2017:3592–5https://doiorg.publicaciones.saludcastillayleon.es/10.1109/embc.2017.8037634
Sennett R, Rendl M. Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling. Seminars Cell Develop Biol. 2012;23:917–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.semcdb.2012.08.011.
Solanas G, Benitah SA. Regenerating the skin: a task for the heterogeneous stem cell pool and surrounding niche. Nature Rev Mol Cell Biol. 2013;14:737–48. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm3675.
Sheen YS, Fan SM, Chan CC, Wu YF, Jee SH, Lin SJ. Visible red light enhances physiological anagen entry in vivo and has direct and indirect stimulative effects in vitro. Lasers in Surg Med. 2015;47:50–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/lsm.22316.
Steward EN, Patel H, Pandya H, Dewan H, Bhavsar B, Shah U, et al. Efficacy of platelet-rich plasma and concentrated growth factor in treating androgenetic alopecia - a retrospective study. Annals of Maxillofac Surg. 2020;10:409–16. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/ams.ams_154_19.
Yuan A, Xia F, Bian Q, Wu H, Gu Y, Wang T, et al. Ceria nanozyme-integrated microneedles reshape the perifollicular microenvironment for androgenetic alopecia treatment. ACS Nano. 2021;15:13759–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsnano.1c05272.
Zhou X, You M, Wang F, Wang Z, Gao X, Jing C, et al. Multifunctional graphdiyne-cerium oxide nanozymes facilitate microrna delivery and attenuate tumor hypoxia for highly efficient radiotherapy of esophageal cancer. Adv Mater (Deerfield Beach, Fla). 2021;33:e2100556. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/adma.202100556.
Zhou D, Du M, Luo H, Ran F, Zhao X, Dong Y, et al. Multifunctional mesoporous silica-cerium oxide nanozymes facilitate miR129 delivery for high-quality healing of radiation-induced skin injury. J Nanobiotechnol. 2022;20:409. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-022-01620-5.
Hang C, Moawad MS, Lin Z, Guo H, Xiong H, Zhang M, et al. Biosafe cerium oxide nanozymes protect human pluripotent stem cells and cardiomyocytes from oxidative stress. J Nanobiotechnol. 2024;22:132. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-024-02383-x.
Mal S, Dwivedi AR, Kumar V, Kumar N, Kumar B, Kumar V. Role of peroxisome proliferator-activated receptor gamma (pparγ) in different disease states: recent updates. Current Med Chem. 2021;28:3193–215. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0929867327666200716113136.
Oyewole AO, Birch-Machin MA. Mitochondria-targeted antioxidants. FASEB J Official Publ Feder Am Soc Exp Biol. 2015;29:4766–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.15-275404.
Lv M, Zhang S, Jiang B, Cao S, Dong Y, Cao L, et al. Adipose-derived stem cells regulate metabolic homeostasis and delay aging by promoting mitophagy. FASEB J Off Publ Feder Am Soc for Exp Biol. 2021;35:e21709. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.202100332R.
Liu Y, Guerrero-Juarez CF, Xiao F, Shettigar NU, Ramos R, Kuan CH, et al. Hedgehog signaling reprograms hair follicle niche fibroblasts to a hyper-activated state. Develop Cell. 2022;57(1758–75):e7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.devcel.2022.06.005.
Ng KJ, Lim J, Tan YN, Quek D, Lim Z, Pantelireis N, et al. Sox2 in the dermal papilla regulates hair follicle pigmentation. Cell Rep. 2022;40:111100. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2022.111100.
Adav SS, Ng KW. Recent omics advances in hair aging biology and hair biomarkers analysis. Ageing Res Rev. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.arr.2023.102041.
Kang Y, Yeo M, Derman ID, Ravnic DJ, Singh YP, Alioglu MA, et al. Intraoperative bioprinting of human adipose-derived stem cells and extra-cellular matrix induces hair follicle-like downgrowths and adipose tissue formation during full-thickness craniomaxillofacial skin reconstruction. Bioact Mater. 2024;33:114–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bioactmat.2023.10.034.
Kruglikov IL, Zhang Z, Scherer PE. The role of immature and mature adipocytes in hair cycling. Trends in Endocrinol Metabol TEM. 2019;30:93–105. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tem.2018.11.004.
Guerrero-Juarez CF, Plikus MV. Emerging nonmetabolic functions of skin fat. Nature Rev Endocrinol. 2018;14:163–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrendo.2017.162.
Nicu C, O’Sullivan JDB, Ramos R, Timperi L, Lai T, Farjo N, et al. Dermal adipose tissue secretes HGF to promote human hair growth and pigmentation. The J Invest Dermatol. 2021;141:1633–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jid.2020.12.019.
Nicu C, Hardman JA, Pople J, Paus R. Do human dermal adipocytes switch from lipogenesis in anagen to lipophagy and lipolysis during catagen in the human hair cycle? Exp Dermatol. 2019;28:432–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.13904.
Tak YJ, Lee SY, Cho AR, Kim YS. A randomized, double-blind, vehicle-controlled clinical study of hair regeneration using adipose-derived stem cell constituent extract in androgenetic alopecia. Stem Cells Trans Med. 2020;9:839–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/sctm.19-0410.
Gertz M, Steegborn C. Using mitochondrial sirtuins as drug targets: disease implications and available compounds. Cell Mol Life Sci: CMLS. 2016;73:2871–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-016-2180-7.
Oláh A, Alam M, Chéret J, Kis NG, Hegyi Z, Szöllősi AG, et al. Mitochondrial energy metabolism is negatively regulated by cannabinoid receptor 1 in intact human epidermis. Exp Dermatol. 2020;29:616–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/exd.14110.
Fields M, Marcuzzi A, Gonelli A, Celeghini C, Maximova N, Rimondi E. Mitochondria-targeted antioxidants, an innovative class of antioxidant compounds for neurodegenerative diseases: perspectives and limitations. Int J Mol Sci. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms24043739.
Mahira Hamdy El Sayed1 MYS, Ahmed Sadek2, Mohamed Abo Shabana Hussein Mohamed3. Efficacy and Tolerability of N-Acetyl-Cysteine for Treatment of The Early-onset Androgenetic Alopecia in Men. An International Journal of Medicine. 2021;114
Yamada Y, Takano Y, Satrialdi AJ, Hibino M, Harashima H. Therapeutic strategies for regulating mitochondrial oxidative stress. Biomolecules. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom10010083.
Huang Y, Ji W, Zhang J, Huang Z, Ding A, Bai H, et al. The involvement of the mitochondrial membrane in drug delivery. Acta Biomater. 2024;176:28–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.actbio.2024.01.027.
Yamada Y, Satrialdi HM, Sasaki D, Abe J, Harashima H. Power of mitochondrial drug delivery systems to produce innovative nanomedicines. Adv Drug Deliver Rev. 2020;154:187–209. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.addr.2020.09.010.
Ganji C, Muppala V, Khan M, Nagaraju GP, Farran B. Mitochondrial-targeted nanoparticles: Delivery and therapeutic agents in cancer. Drug Discove Today. 2023;28:103469. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.drudis.2022.103469.
Tabish TA, Lygate CA. Mitochondria-targeted nanomedicines for cardiovascular applications. Nanomedicine. 2023;18:2101–4. https://doiorg.publicaciones.saludcastillayleon.es/10.2217/nnm-2023-0321.
Yamada Y, Munechika R, Satrialdi KF, Sato Y, Sakurai Y, et al. Mitochondrial delivery of an anticancer drug via systemic administration using a mitochondrial delivery system that inhibits the growth of drug-resistant cancer engrafted on mice. J Pharmaceu Sci. 2020;109:2493–500. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.xphs.2020.04.020.
Liu Y, Zhang J, Tu Y, Zhu L. Potential-independent intracellular drug delivery and mitochondrial targeting. ACS Nano. 2022;16:1409–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsnano.1c09456.
Davis MG, Piliang MP, Bergfeld WF, Caterino TL, Fisher BK, Sacha JP, et al. Scalp application of antioxidants improves scalp condition and reduces hair shedding in a 24-week randomized, double-blind, placebo-controlled clinical trial. Int J Cosmet Sci. 2021;43(1):14–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ics.12734.
Davis MG, Piliang MP, Bergfeld WF, Caterino TL, Fisher BK, Sacha JP, et al. Scalp application of the antioxidant piroctone olamine reduces hair shedding in an 8-week randomized, double-blind, placebo-controlled clinical study. Int J Cosmet Sci. 2021;43(Suppl 1):S26–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ics.12737.
Liu D, Gao Y, Liu J, Huang Y, Yin J, Feng Y, et al. Intercellular mitochondrial transfer as a means of tissue revitalization. Signal Transduct Target Ther. 2021;6:65. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-020-00440-z.
Ramos PM, Brianezi G, Martins ACP, Silva MGD, Marques MEA, Miot HA. Aryl hydrocarbon receptor overexpression in miniaturized follicles in female pattern hair loss. An Bras Dermatol. 2017;92:430–1. https://doiorg.publicaciones.saludcastillayleon.es/10.1590/abd1806-4841.20175150.
Ramos PM, Brianezi G, Martins AC, da Silva MG, Marques ME, Miot HA. Apoptosis in follicles of individuals with female pattern hair loss is associated with perifollicular microinflammation. Int J Cosmet Sci. 2016;38:651–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ics.12341.
Sahebnasagh A, Hashemi J, Khoshi A, Saghafi F, Avan R, Faramarzi F, et al. Aromatic hydrocarbon receptors in mitochondrial biogenesis and function. Mitochondrion. 2021;61:85–101. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mito.2021.09.012.
Acknowledgements
The authors declare that they have not used AI-generated work in this manuscript.
Funding
This work was supported by the Zhejiang Provincial Natural Science Foundation of China (grant No. LY21H110001), the Zhejiang Medical and Health Science and Technology Project (grant No. 2021KY903), and the Hangzhou Medical Key Discipline Construction Project (grant No. [2021]21–3).
Author information
Authors and Affiliations
Contributions
Tingru Dong contributed to writing specific sections of the manuscript and creating all illustrations. Yujie Li drafted the remaining sections. Shiyu Jin, Fenglan Yang, and Renxue Xiong were responsible for collecting references. Yeqin Dai offered advice on writing. Xiuzu Song provided funding support. Cuiping Guan supervised the overall design and revision of the manuscript, as well as providing funding support. All authors have thoroughly reviewed and unanimously approved the final version of the manuscript.
Corresponding author
Ethics declarations
Ethical approval and consent to participate
Not applicable
Consent for publication
Not applicable
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Dong, Tr., Li, Yj., Jin, Sy. et al. Progress on mitochondria and hair follicle development in androgenetic alopecia: relationships and therapeutic perspectives. Stem Cell Res Ther 16, 44 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04182-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04182-z