Skip to content
BY 4.0 license Open Access Published by De Gruyter April 5, 2023

Male-pattern hair loss: Comprehensive identification of the associated genes as a basis for understanding pathophysiology

  • Sabrina K. Henne

    Sabrina K. Henne, MSc

    ORCID logo
    , Markus M. Nöthen

    Prof. Dr. med. Markus M. Nöthen

    ORCID logo
    and Stefanie Heilmann-Heimbach

    PD Dr. rer. nat. Stefanie Heilmann-Heimbach

    ORCID logo EMAIL logo
From the journal Medizinische Genetik

Abstract

Male-pattern hair loss (MPHL) is a highly heritable and prevalent condition that is characterized by progressive hair loss from the frontotemporal and vertex scalp. This androgen-dependent hair loss may commence during puberty, and up to 80 % of European men experience some degree of MPHL during their lifetime. Current treatment options for MPHL have limited efficacy, and improved understanding of the underlying biological causes is required to facilitate novel therapeutic approaches. To date, molecular genetic studies have identified 389 associated genomic regions, have implicated numerous genes in these regions, and suggested pathways that are likely to contribute to key pathophysiological mechanisms in MPHL. This review provides an overview of the current status of MPHL genetic research. We discuss the most significant achievements, current challenges, and anticipated developments in the field, as well as their potential to advance our understanding of hair (loss) biology, and to improve hair loss prediction and treatment.

Introduction

Male pattern hair loss (MPHL), also termed male androgenetic alopecia, is a highly heritable and age-dependent trait. The hair loss may commence during puberty and has a lifetime prevalence of ~80 % in European men [1]. The phenotype is characterized by a distinct pattern of progressive, age-dependent hair loss from the scalp. MPHL typically begins with a bi-temporal recession of the frontal hair line, followed by a thinning of hair in the frontal and vertex scalp areas, which eventually results in complete baldness of the top of the scalp (Figure 1) [2]. In addition to this characteristic pattern of hair loss, the key pathophysiological features of MPHL include a strict androgen-dependency of the phenotype; changes in hair cycle dynamics (i. e. a shorter growth phase [anagen] and a prolonged resting phase [telogen]); and the miniaturization of affected hair follicles (HFs), causing a transition of pigmented terminal hairs to unpigmented vellus hairs [3].

Figure 1: Clinical classification of MPHL according to the Hamilton-Norwood scale. MPHL is classified clinically into seven major stages, starting from stage I (no hair loss), a receding of the fronto-temporal hairline (II), regression of the frontal hairline and balding of frontal and vertex scalp areas (III–IV), confluence of the affected areas (V–VI) until only an occipitotemporal hair crown persists (VII) [4]. The less common a variant of MPHL progression (IIa, IIIa, IVa, Va) is characterized by recession of the entire anterior hairline without simultaneous development of a bald area on the vertex.
Figure 1:

Clinical classification of MPHL according to the Hamilton-Norwood scale. MPHL is classified clinically into seven major stages, starting from stage I (no hair loss), a receding of the fronto-temporal hairline (II), regression of the frontal hairline and balding of frontal and vertex scalp areas (III–IV), confluence of the affected areas (V–VI) until only an occipitotemporal hair crown persists (VII) [4]. The less common a variant of MPHL progression (IIa, IIIa, IVa, Va) is characterized by recession of the entire anterior hairline without simultaneous development of a bald area on the vertex.

MPHL – especially when occurring in early adolescence – can exert profound negative effects on quality of life [5]. Current therapeutic options for MPHL are limited. At writing, only two U.S. Food and Drug Administration-approved drugs are available, i. e. oral finasteride and topical minoxidil. These vary in terms of efficacy, are non-curative, and can induce severe side effects [3], [6]. The development of novel therapeutic approaches to MPHL will require an improved understanding of the underlying biological causes. This in turn requires the elucidation of the underlying genetic basis. To date, molecular genetic studies of MPHL have identified 389 risk loci, and have led to the identification of pathobiologically relevant genes and pathways for MPHL. This review provides an overview of the key findings, current challenges, and anticipated developments in the field of MPHL genetic research. We also discuss the potential of these developments in terms of advancing our understanding of hair (loss) biology, facilitating drug target identification, and developing new therapies, and in elucidating MPHL pathobiology and its shared biological basis with other traits and diseases.

MPHL – a highly heritable and polygenic trait

The familial occurrence of MPHL is well documented [7]. While in early research, the pattern of familial occurrence was thought to be compatible with a simple Mendelian mode of inheritance [8], later authors proposed a polygenic model involving a multitude of genes [9], [10] and this hypothesis is now considered proven. Two early twin studies estimated the heritability (h2) of MPHL as 0.81 (95 % confidence interval [CI] 0.77–0.85) in early-onset MPHL families (25–36 years), and 0.79 (95 % CI 0.4–0.85) in elderly males (> 70 years), respectively [11], [12]. However, the true heritability may be even higher, since misclassifications of MPHL-severity (e. g. through classification by self-assessment) would lead to an underestimation of MPHL heritability [3]. The strongly heritable and polygenic nature of MPHL was confirmed in a pedigree-based h2 analysis in pairs of first-degree relatives from the UK Biobank (UKB) cohort, which generated an h2 estimate of 0.62 [13]. The same study found that approximately 60 % of the genetic contribution to MPHL variance was captured by common single nucleotide polymorphisms (SNPs) (h2SNP = 0.393), and that the additional inclusion of rare SNPs (1.5x10–5 < minor allele frequency [MAF] ≤ 1x10–3) had only minor effects on h2 estimates (h2SNP = 0.415). Of note, pedigree-based h2 estimates were slightly lower for father-son correlations compared to those for brother-brother pairs. This may in part reflect the X-chromosome contribution, since fathers and sons do not share X-chromosomal risk factors by descent. One other potential explanation for this observation is that during childhood, brothers share a common environment. However, the issue of whether – and if so to what extent – environmental factors (e. g. smoking, alcohol consumption) are implicated in MPHL development remains a matter of debate [14]–[20].

Findings from 25 years of gene identification efforts

Since 1998, multiple molecular genetic studies of MPHL have been performed to identify causal genes and pathways. Early MPHL genetic studies were limited to the investigation of only a few, or even single, genes, and generated limited insights into the genetic basis of MPHL. One important caveat here, however, is that even negative findings could not exclude a genuine genetic contribution for the respective gene, since the investigated sample sizes were limited.

A key exception is the X-linked androgen receptor gene (AR), which became the focus of a number of studies due to the strict androgen dependency of the MPHL phenotype [21]–[29]. Although these studies generated contradictory results in terms of the associated variants and the most likely causal gene in the region, the genomic region surrounding the AR is without question the most strongly associated region for MPHL in the human genome. However, unequivocal identification of the precise causative variants at the X-chromosomal AR/EDA2R locus has not yet been achieved, and neither AR nor the neighbouring EDA2R have been confirmed as the causal gene.

Following these investigations, no notable progress in the identification of additional genetic risk factors was then made until the publication of the first two genome-wide association studies (GWAS) of MPHL in 2008. Since then, GWAS have been performed in increasingly large cohorts, and have generated substantial insights into the role of common genetic variants in MPHL susceptibility (Figure 2). At writing, the GWAS Catalog [30] lists a total of 13 studies under the terms “androgenetic alopecia” or “balding measurement”, 10 of which report data on MPHL (Table 1). These studies can be broadly categorized into single cohort GWAS, international meta-analyses, and population-based GWAS. The number of reported association signals per study strongly correlates with sample size, and ranges from two independent risk variants in the first published GWAS studies [31], [32] to 622 independent risk variants in the largest GWAS to date [13]. While the individual studies used varying definitions to classify independent risk loci, collapsing of the reported association signals into independent risk loci based on the FUMA tool [33] results in a total of 389 known risk loci for MPHL. Together, these loci explain ~39 % of the observed phenotypic variance in MPHL [13].

Figure 2: MPHL susceptibility loci identified via GWAS of individuals of European descent between 2008 and 2022. MPHL genetic studies are represented by circles. The number of loci denotes the cumulative number of independent MPHL susceptibility loci. The number of individuals and the number of cases in the cohort are reflected by the sizes of the grey and blue circles, respectively. Reference numbers are given in square brackets.
Figure 2:

MPHL susceptibility loci identified via GWAS of individuals of European descent between 2008 and 2022. MPHL genetic studies are represented by circles. The number of loci denotes the cumulative number of independent MPHL susceptibility loci. The number of individuals and the number of cases in the cohort are reflected by the sizes of the grey and blue circles, respectively. Reference numbers are given in square brackets.

The vast majority of these loci are located on the autosomes, and are therefore equally likely to have been inherited from the maternal or the paternal side. Six risk loci, however, are located on the maternally inherited X-chromosome. These include the AR/EDA2R locus, which has been confirmed as the most strongly associated genomic locus across all studies [13], [31], [32], [34]–[39]. The available data suggest that maternal heredity plays a greater role in the inheritance of MPHL risk than paternal heredity. However, no studies have yet been performed to investigate the existence of paternally inherited Y-chromosomal risk factors.

From association signals to candidate genes and disease mechanisms

The majority of MPHL risk variants are located in non-coding – and mostly intergenic – regions of the genome. A reasonable assumption is that these variants exert regulatory effects on the expression of relevant genes, e. g. through changes in transcription factor binding sites or enhancer/promoter interaction [41]. While in early GWAS, the proximity of a gene to an association signal was considered the strongest indicator of disease relevance, a number of bioinformatic tools have since been developed that prioritize disease genes and pathways from GWAS data using multiple lines of evidence [33], [42]. The application of these tools to MPHL genetic data has resulted in the identification of a number of interesting genes and pathways (Figure 3). These include the genes encoding i) steroid-5-α-reductase type II (SRD5A2), which is the only known therapeutic target for MPHL; ii) WNT10A and fibroblast growth factor 5 (FGF5), which are known regulators of hair cycling and hair growth, respectively; iii) interferon regulatory factor 4 (IRF4), which has previously been reported in connection with hair pigmentation; iv) the transcriptional regulators early B cell factor 1 (EBF1), TWIST1, and TWIST2; and v) other genes involved in the WNT signalling pathway (DKK2–Dickkopf 2, FZD10–Frizzled class receptor 10, FAM53B) or androgen signalling (e. g. TOP1, FAM9B, HDAC4, and HDAC9) [3], [38]. Pathway-based and gene-set enrichment analyses have underlined the importance of androgen- and WNT-signalling in MPHL pathogenesis, and have also yielded evidence for a number of other hormonal pathways (e. g. oestrogen- and melatonin signalling, reviewed elsewhere [43]), as well as pathways involved in skin- and epidermal development, apoptosis, and adipogenesis, and immunological processes [13], [38], [39]. Besides indicating the relevance of cells and processes within the HF per se, the latter implicate the perifollicular environment, such as the resident immune cells and adipocytes of the scalp [38].

In studies of MPHL risk variants, regulatory effects on candidate gene expression have been reported at four GWAS loci (1p12–FAM46C; 2q35–WNT10A; 15q22.2–RORA; 21q22.1–RCAN1) [44], [45]. Among these, the most notable regulatory effect was found for the MPHL risk allele rs7349332-T at 2q35, which resulted in reduced WNT10A expression in human HFs [36]. A follow-up study to investigate the underlying molecular mechanism at this locus identified a nearby binding site for the transcription factor EBF1, whose encoding gene is located at a second MPHL risk locus at 5q33.3. The study demonstrated that EBF1 activates the WNT10A promoter, and that this interaction is impacted by the allelic expression of the 2q35 MPHL risk variant. These findings indicate a functional interaction between genes at two independent MPHL risk loci, and suggest that the 2q35 MPHL risk allele results in decreased WNT10A promoter activation via EBF1 and a resultant lowering of WNT10A expression, which eventually impacts HF cycling [46].

Table 1:

Overview of GWAS, number of reported independent associations (P < 5x10–8), and key findings to date. When available, the number of samples and cases is shown.

Authors

Year

Study type

N samples (cases)

N associations

(P < 5x10–8)

Key findings

Hillmer et al.

[31]

2008

GWAS

643 (296)

2

Confirmation of the AR/EDA2R locus, novel risk locus on chr20p11.22 (concurrent with [32])

Richards et al.

[32]

2008

GWAS

1125 (578)

2

Confirmation of the AR/EDA2R locus, novel risk locus on chr20p11.22 (concurrent with [31])

Brockschmidt et al. [34]

2011

GWAS

1198 (581)

3

Novel risk locus on chr7p21.1, implicating HDAC9

Li et al.

[35]

2012

Meta-analysis

12,806 (3,891)

8

1st international meta-analysis of the MAAN consortium. Identification of 5 novel autosomal risk loci, unexpected association with Parkinson’s disease

Heilmann et al.

[36]

2013

Candidate variant analysis

5,420 (2,759)

4

First genetic evidence for an involvement of WNT-signalling (WNT10A, 2q35) in MPHL

Pickrell et al.

[40]

2016

GWAS

17,500 (9,009)

49

Evidence for pleiotropic effects of MPHL risk variants on lower age at menarche

Hagenaars et al. [37]

2017

Population-based GWAS

52,874 (36,150)

287

1st GWAS based on data from the UK Biobank, development of a polygenic prediction model

Heilmann-Heimbach et al. [38]

2017

Meta-analysis

22,518 (10,846)

63

2nd MAAN analysis, implication of numerous candidate genes and pathways, evidence for shared genetic basis between MPHL and other traits and illnesses

Pirastu et al.

[39]

2017

Population-based GWAS

43,590 (25,662)

71

Implication of plausible additional candidate genes and pathways (e. g. TGF-beta, apoptosis signalling)

Yap et al.

[13]

2018

Population-based GWAS

205,327

622

Largest GWAS to date

While the underlying genes and precise biological mechanisms at the majority of loci remain elusive, for some loci and genes, hypotheses can be formulated as to how they will contribute to key pathophysiological processes in MPHL. These include deregulation of the hair growth cycle (FGF5, EBF1, DKK2, adipogenesis, WNT-signalling), mediation of androgen sensitivity (SRD5A2, AR-signalling, melatonin signalling), and the conversion of pigmented terminal into non-pigmented vellus-like hairs (IRF4) (Figure 3).

Epidemiological associations and genetic pleiotropy – placing MPHL into a wider medical context

Epidemiological studies have reported associations between MPHL and a number of medical conditions and anthropometric indices, including cardiovascular disease [47]–[54], type 2 diabetes [18], [53], [55]–[58], BMI [18], [58], [59], metabolic syndrome [58], [60]–[63], benign prostate hyperplasia [64], [65] and prostate cancer [66], [67]. The observed epidemiological associations suggest an overlap in pathobiology, which may be explained at least in part by shared genetic factors. Using GWAS data, genome-wide significant correlations have been detected for MPHL and metrics of early puberty [13], [40], bone mineral density (↑) [13], HOMA-B levels (↑) [13] and human body height (↓) [39]. Since early puberty is associated with childhood adiposity and increased BMI in adulthood [68], and higher HOMA-B levels (a measure of pancreatic β-cell function), are associated with an increased BMI [69], these genetic correlations may partly explain the reported epidemiological association between MPHL and metabolic syndrome, and between MPHL and BMI [13], [39]. At individual loci, overlapping associations have been reported for prostate cancer and cardiovascular traits, although these did not reflect in a significant genome-wide genetic correlation [13], [18], [37], [40].

Figure 3: From genetic association finding to an understanding of MPHL pathobiology. GWAS have identified 389 independent risk loci and implicated numerous candidate genes and pathways. In some cases, assumptions can be made as to how these genes and pathways contribute to the key pathophysiological features that result in the characteristic MPHL phenotype. Inside to outside: Genomic location (chromosome 1–chromosome X) of candidate genes and assignment to pathways; Manhattan plot of genome-wide significant MPHL SNPs [13] (Y-Axis corresponds to 7–150-log10(P)); key pathophysiological features of MPHL. Colour coding represents the assignment of genes to pathways and their contribution to MPHL key pathophysiological features.
Figure 3:

From genetic association finding to an understanding of MPHL pathobiology. GWAS have identified 389 independent risk loci and implicated numerous candidate genes and pathways. In some cases, assumptions can be made as to how these genes and pathways contribute to the key pathophysiological features that result in the characteristic MPHL phenotype. Inside to outside: Genomic location (chromosome 1–chromosome X) of candidate genes and assignment to pathways; Manhattan plot of genome-wide significant MPHL SNPs [13] (Y-Axis corresponds to 7–150-log10(P)); key pathophysiological features of MPHL. Colour coding represents the assignment of genes to pathways and their contribution to MPHL key pathophysiological features.

A study in the Icelandic population found a pleiotropic variant for MPHL and reduced female reproductive rate on ch.17q21.31 [35], [70]. Notably, polycystic ovary syndrome (PCOS), the most common cause of reduced fertility in women, is frequently observed in the female relatives of MPHL-affected men [71], [72], and a Mendelian randomization study found that MPHL risk variants are associated with increased PCOS risk [73]. However, a genome-wide genetic correlation analysis found no significant correlation between the two traits [73].

At single loci, overlapping genetic associations with MPHL have also been reported for Parkinson’s disease (PD) and amyotrophic lateral sclerosis (ALS) [35]. These findings were supported by subsequent epidemiological investigations, which revealed an increased risk for PD (1.3-fold increased risk; 95 % CI 1.06–1.55 [35]) and ALS (2.7-fold increased risk; 95 % CI 1.23–6.31 [74]) in MPHL-affected compared to unaffected men. However, no genome-wide genetic correlations were observed.

More recently, research has suggested that MPHL is a possible risk factor for severe COVID-19, as based on the high incidence of severe MPHL in hospitalized COVID-19 patients [75]. However, subsequent epidemiological and genetic correlation analyses did not confirm this epidemiological association [76]–[78], and no general genetic correlation was identified [76], [79], although pathway-based polygenic risk score analyses suggested a potential limited biological overlap between the traits [76].

In many cases, steroid hormone or androgen signalling may act as the common biological mechanism for MPHL and the associated traits. Androgen-mediated neurotoxicity has been discussed as a possible mechanism in the pathogenesis of PD, and could also partially explain the association with ALS [80], [81]. In women, PCOS is promoted by elevated androgen levels [82]. Androgen- and oestrogen-signalling, together with other MPHL-associated hormonal pathways, have also been implicated in skeletal development and bone homeostasis [83], [84].

Future studies are required to determine the extent to which these pleiotropies can indeed be explained by common hormonal pathways, and to identify other contributory biological signalling pathways. A deeper understanding of pleiotropies and the underlying biological mechanisms will have important implications for risk prediction and drug therapy, both in terms of drug repurposing and the prediction of side effects.

To lose or not to lose – MPHL risk predicted from genetic data

Although several MPHL prediction models have been developed, to date, these have yielded only limited predictive value at the individual level [37], [85]–[87]. The most recent model was based on 160,267 male participants from the UKB, with testing having been performed in an additional 26,177 male UKB participants as well as in an independent cohort of 991 males enriched for early-onset (< 40 years) severe MPHL cases [87]. When predicting MPHL with differing degrees of severity, the model achieved prediction accuracies ranging from 0.60 – 0.73, as based on the area under the curve (AUC), with higher degrees of severity being predicted with greater accuracy. A prediction accuracy of 0.83 was achieved when predicting the presence of any hair loss in the early-onset (< 40 years) cohort. Notably, the strongest predictor was age, followed by rs12558842, which is located ~280kb upstream of AR [87]. Although the overall prediction accuracy and reliability of the model have improved substantially over time, the accuracy of individual predictions remains limited, in particular for mild and medium levels of balding (Hamilton-Norwood-scale < VI). Prediction may therefore be too crude for reliable estimates of metrics such as an individual’s age of onset or their rate of MPHL progression. While such models could now be expanded due to the discovery of novel risk loci and variants [13], further investigations of additional genetic risk factors and potential environmental factors, and the use of large independent data sets, will also be required to improve current MPHL prediction models. Moreover, existing prediction models are based on Euro-centric GWAS, which will likely hamper their application in other ethnicities [88]. Once successfully established, MPHL prediction models may be valuable in a (wider) medical and forensic context. Prediction could also help to determine the feasibility and appropriate time for drug interventions and hair transplantations, for which therapeutic efficiency is strongly dependent on the clinical stage and the rate of disease progression [6], [89].

MPHL genetic studies in non-European ethnicities

Epidemiological research indicates that MPHL manifests slightly differently in non-European ethnicities in terms of prevalence, onset, and pattern of progression [1], [90]–[92]. However, most genetic studies to date have focused on Europeans, with genetic studies in non-European populations having been limited to one GWAS of combined female-pattern hair loss (FPHL) and MPHL in admixed Latin Americans [93], two replication studies in Asian populations [94], [95], and one small-scale autosomal GWAS of MPHL in a cohort from Korea [95]. The Latin American GWAS identified associations at the AR/EDA2R locus, with the most significant variant rs4258142 having been previously described in Europeans, and rs2814331 at the 10q23.2 locus implicating GRID1 as a novel candidate gene [93]. However, since MPHL and FPHL were jointly analysed, it is unclear whether this association was attributable to MPHL. The Korean-based GWAS identified 13 suggestive (P < 10–5) risk loci (1p21.1, 4q22.3, 4q35.1, 5p15.33, 7q31.31, 9q21.31, 9q21.33, 10p11.21, 10q23.31, 11q21, 11q25, 12p11.21, 18p11.21), one of which (9q21.31) had been described in a previous European-based GWAS. The authors also investigated SNPs with a previously reported association with MPHL in European-based GWAS. The strongest nominally significant (P < 0.05) associations were found at 20p11 [95], as in a previous replication attempt in the Chinese Han population [94].

Notably, the association at the AR/EDA2R locus could not be replicated in the Asian studies, since previously implicated SNPs at the locus were monomorphic or nearly monomorphic (MAFs < 1 %) for the European MPHL risk alleles in the Korean and Chinese Han populations [94], [95]. This may either point to a different allelic architecture and/or haplotype constellation between European and Asian populations at the AR/EDA2R locus or a general genetic heterogeneity for MPHL between these populations. Independent analyses of the X chromosome are therefore required to clarify the role of variants at the AR/EDA2R locus in Asian populations. While the association findings in non-European populations require replication in larger cohorts, they suggest the presence of distinct risk loci in non-European ethnicities. Further studies in diverse populations are required to shed light on genetic determinants in other ethnicities. This could facilitate further exploration of MPHL pathobiology and the identification of mechanisms that contribute to ethnic differences, such as age of onset and balding severity, in specific subpopulations of androgen-sensitive scalp HFs.

Overlap between MPHL and female-pattern hair loss

The present review has focused on the advances and challenges of genetic research into MPHL, which raises the question of the extent to which these findings also apply to FPHL. The two conditions share key characteristics, such as high prevalence, the age-dependent and progressive nature of the hair loss, and key pathophysiological characteristics, such as follicular miniaturization and deregulation of hair cycle dynamics [2], [96]. However, research suggests that MPHL and FPHL are distinct entities with differing pathogeneses [97], and the relevance of genetic factors and the role of androgens in FPHL is less clear than is the case for MPHL [96], [97]. While previous authors have suggested that FPHL has a heritable component [98], [99], the evidence is inconclusive [97]. Genetic studies of FPHL have largely focused on specific genes related to steroid hormone metabolism and on risk variants and loci previously identified in MPHL. Together, these studies have implicated a number of candidate genes (CYP19A1, ESR2, PPARGC1A, ABCC4, CYP11B2, FSHB) and risk loci (AR/EDA2R, 20p11) [97], [100] – [111]. To date, no GWAS of FPHL has yet been published. GWAS in larger FPHL cohorts are required to improve estimation of the contribution of genetic factors, and to generate novel insights into the underlying genetic architecture.

Challenges and perspectives in MPHL genetic research

MPHL is widely misconceived as a phenotype of mainly cosmetic and psychological importance. However, given its high prevalence, simple clinical classification, strict hormone dependency, sex-limited expression, and highly polygenic basis, MPHL is an interesting model phenotype for the study of traits with similar features.

Available GWAS of MPHL have identified associations with thousands of common variants at 389 loci, and have yielded unprecedented insights into the genetic and biological basis of what is the most common form of hair loss in men. Together, these genetic findings explain ~39 % of the observed phenotypic variance of MPHL in the white British population [13], [39], mixed European populations [38], and Latin American populations [93]. Research has confirmed that MPHL has a strong genetic contribution, with SNP-based h2 estimates of 60–70 % [13]. However, research has yet to demonstrate the existence of genetic factors that may bridge the gap between the SNP-based and the twin-study h2 estimates of MPHL heritability. While a general hypothesis is that rare genetic variants contribute to complex disease heritability, their estimated contribution to MPHL is minimal [13], and to date, exome-based phenome-wide association studies (PheWAS) have identified no associations between rare variants and MPHL [112], [113]. Despite the fact that rare variants are unlikely to contribute in general to MPHL heritability, the possibility that they play a role in some individuals and families cannot be excluded. However, in contrast to common variants, few data are available concerning the contribution of individual rare variants (MAF < 1 %) to MPHL in phenotypically well characterized or family-based cohorts. Each of these rare genetic findings will add important information on the genotype-phenotype correlation, and may provide important (new) insights into MPHL pathobiology. Future genetic studies are warranted to decipher the full allelic spectrum of MPHL, and these should include larger cohorts and the investigation of the full spectrum (common and rare variants) of sequence variation through whole-exome or whole-genome sequencing. Comprehensive understanding of MPHL phenotypic variance in the population will require the investigation of the genetic – and potentially also the non-genetic factors – that impact hair loss progression. This in turn will require longitudinal studies in large cohorts with extensive phenotypic and comprehensive genetic data. Furthermore, future systematic investigations into the genetic basis of MPHL both within different ethnicities and across different ethnicities will yield further insights into MPHL pathobiology, and will enable the generation of improved and trans-ethnic prediction models.

In addition to the further dissection of the genetic basis of MPHL, another important and likely even greater challenge will be the functional annotation and biological interpretation of current and future genetic findings. Although several plausible genes and pathways have been identified, the detection of the causal genes and pathways, and our understanding of the precise underlying biological mechanisms, remain in their infancy. Despite the fact that the functional annotation of non-coding regions in the genome is becoming increasingly detailed [114], further experiments are required to accurately reflect the full spectrum of regulatory interactions, which are often tissue-, cell type-, or context-specific. This is also true for MPHL, for which only a limited number of human data sets are available for the identification of causal genes and pathways and the investigation of the molecular mechanisms that underlie the characteristic hair loss.

Systematic follow-up of all association findings for MPHL is warranted, and this is likely to involve two key approaches. First, the development of sophisticated bioinformatic analysis strategies will probably be required to allow the performance of pathway- and network-based analyses that elucidate the molecular mechanisms at individual and across loci. Second, (single-cell) expression profiling of genes and signalling pathways will probably be necessary in order to determine the relevant HF and/or perifollicular compartments in which known, and as yet unidentified, pathways interact to cause the characteristic androgen-dependent hair loss.

As discussed, current therapeutic options for MPHL are suboptimal, and knowledge of MPHL relevant genes and pathways will facilitate the development of further (more effective) therapies. In a first step towards this, work by Pirastu et al. and our group showed that 45 MPHL candidate genes were druggable targets [39, own unpublished data]. These findings are encouraging, since drugs based on targets or mechanisms for which genetic evidence is available are more likely to succeed in clinical trials [115], [116]. Furthermore, due to the highly polygenic nature of MPHL, genetically guided drug research and patient stratification may eventually lead to the accurate prediction of therapeutic success and adverse side effects, thus enabling precision medicine approaches.

About the authors

Sabrina K. Henne

Sabrina K. Henne, MSc

Prof. Dr. med. Markus M. Nöthen PD

Prof. Dr. med. Markus M. Nöthen

PD Dr. rer. nat. Stefanie Heilmann-Heimbach

PD Dr. rer. nat. Stefanie Heilmann-Heimbach

References

[1] J. B. Hamilton, “Patterned loss of hair in man; types and incidence,” Ann N Y Acad Sci, vol. 53, no. 3, pp. 708–728, Mar. 1951, doi: 10.1111/j.1749-6632.1951.tb31971.x.10.1111/j.1749-6632.1951.tb31971.xSearch in Google Scholar PubMed

[2] J. Qi and L. A. Garza, “An overview of alopecias,” Cold Spring Harb Perspect Med, vol. 4, no. 3, 2014, doi: 10.1101/cshperspect.a013615.10.1101/cshperspect.a013615Search in Google Scholar PubMed PubMed Central

[3] S. Heilmann-Heimbach, L. M. Hochfeld, R. Paus, and M. M. Nöthen, “Hunting the genes in male-pattern alopecia: how important are they, how close are we and what will they tell us?,” Exp Dermatol, vol. 25, no. 4, pp. 251–257, Apr. 2016, doi: 10.1111/exd.12965.10.1111/exd.12965Search in Google Scholar PubMed

[4] O. T. Norwood, “Male pattern baldness: Classification and incidence,” South Med J, vol. 68, no. 11, pp. 1359–1365, 1975, doi: 10.1097/00007611-197511000-00009.10.1097/00007611-197511000-00009Search in Google Scholar PubMed

[5] N. L. Yu, H. Tan, Z. Q. Song, and X. C. Yang, “Illness perception in patients with androgenetic alopecia and alopecia areata in China,” J Psychosom Res, vol. 86, pp. 1–6, Jul. 2016, doi: 10.1016/J.JPSYCHORES.2016.04.005.10.1016/j.jpsychores.2016.04.005Search in Google Scholar PubMed

[6] D. Rathnayake and R. Sinclair, “Male androgenetic alopecia,” Expert Opin Pharmacother, vol. 11, no. 8, pp. 1295–1304, Jun. 2010, doi: 10.1517/14656561003752730.10.1517/14656561003752730Search in Google Scholar PubMed

[7] H. Davis, “Cases of Inherited Alopecia,” http://dx.doi.org/10.1177/003591571300600303, vol. 6, no. Dermatol_Sect, p. 6, Sep. 2016, doi: 10.1177/003591571300600303.10.1177/003591571300600303Search in Google Scholar

[8] D. Osborn, “Inheritance of baldness: Various patterns due to heredity and sometimes present at birth-a sex-limited character-dominant in man-women not bald unless they inherit tendency from both parents,” Journal of Heredity, vol. 7, no. 8, pp. 347–355, Aug. 1916, doi: 10.1093/OXFORDJOURNALS.JHERED.A110746/2/7-8-347.PDF.GIF.10.1093/oxfordjournals.jhered.a110746Search in Google Scholar

[9] W. Küster and R. Happle, “The inheritance of common baldness: Two B or not two B?,” J Am Acad Dermatol, vol. 11, no. 5, pp. 921–926, Nov. 1984, doi: 10.1016/S0190-9622(84)80498-3.10.1016/S0190-9622(84)80498-3Search in Google Scholar PubMed

[10] S. Heilmann et al., “Evidence for a polygenic contribution to androgenetic alopecia,” British Journal of Dermatology, vol. 169, no. 4, pp. 927–930, Oct. 2013, doi: 10.1111/BJD.12443.10.1111/bjd.12443Search in Google Scholar PubMed

[11] D. R. Nyholt, N. A. Gillespie, A. C. Heath, and N. G. Martin, “Genetic Basis of Male Pattern Baldness,” Journal of Investigative Dermatology, vol. 121, no. 6, pp. 1561–1564, Dec. 01, 2003, doi: 10.1111/j.1523-1747.2003.12615.x.10.1111/j.1523-1747.2003.12615.xSearch in Google Scholar PubMed

[12] H. Rexbye et al., “Hair Loss Among Elderly Men: Etiology and Impact on Perceived Age,” J Gerontol A Biol Sci Med Sci, vol. 60, no. 8, pp. 1077–1082, Aug. 2005, doi: 10.1093/gerona/60.8.1077.10.1093/gerona/60.8.1077Search in Google Scholar PubMed

[13] C. X. Yap et al., “Dissection of genetic variation and evidence for pleiotropy in male pattern baldness,” Nat Commun, vol. 9, no. 1, Dec. 2018, doi: 10.1038/s41467-018-07862-y.10.1038/s41467-018-07862-ySearch in Google Scholar PubMed PubMed Central

[14] M. Danesh-Shakiba, J. Poorolajal, and P. Alirezaei, “Androgenetic Alopecia: Relationship to Anthropometric Indices, Blood Pressure and Life-Style Habits,” 2020, doi: 10.2147/CCID.S231940.10.2147/CCID.S231940Search in Google Scholar PubMed PubMed Central

[15] Y. Kavadya and V. Mysore, “Role of smoking in androgenetic alopecia: A systematic review,” International Journal of Trichology, vol. 14, no. 2. Wolters Kluwer Medknow Publications, pp. 41–48, Mar. 01, 2022. doi: 10.4103/ijt.ijt_59_21.10.4103/ijt.ijt_59_21Search in Google Scholar PubMed PubMed Central

[16] A. Babadjouni, D. Pouldar Foulad, B. Hedayati, E. Evron, and N. Mesinkovska, “The Effects of Smoking on Hair Health: A Systematic Review,” Skin Appendage Disorders, vol. 7, no. 4. S. Karger AG, pp. 251–264, Jun. 01, 2021. doi: 10.1159/000512865.10.1159/000512865Search in Google Scholar PubMed PubMed Central

[17] R. M. Trüeb, “Effect of ultraviolet radiation, smoking and nutrition on hair,” Current Problems in Dermatology (Switzerland), vol. 47, pp. 107–120, 2015, doi: 10.1159/000369411.10.1159/000369411Search in Google Scholar PubMed

[18] S. Pechlivanis et al., “Male-pattern baldness and incident coronary heart disease and risk factors in the Heinz Nixdorf Recall Study,” PLoS One, vol. 14, no. 11, Nov. 2019, doi: 10.1371/journal.pone.0225521.10.1371/journal.pone.0225521Search in Google Scholar PubMed PubMed Central

[19] K. Esen Salman, N. A. Kucukunal, I. Kivanc Altunay, and A. Aksu Cerman, “Frequency, severity and related factors of androgenetic alopecia in dermatology outpatient clinic: Hospital-based cross-sectional study in Turkey,” An Bras Dermatol, vol. 92, no. 1, pp. 35–40, Jan. 2017, doi: 10.1590/abd1806-4841.20175241.10.1590/abd1806-4841.20175241Search in Google Scholar PubMed PubMed Central

[20] G. Severi et al., “Androgenetic alopecia in men aged 40–69 years: Prevalence and risk factors,” British Journal of Dermatology, vol. 149, no. 6, pp. 1207–1213, Dec. 2003, doi: 10.1111/j.1365-2133.2003.05565.x.10.1111/j.1365-2133.2003.05565.xSearch in Google Scholar PubMed

[21] F. F. Brockschmidt et al., “Fine mapping of the human AR/EDA2R locus in androgenetic alopecia,” Br J Dermatol, vol. 162, no. 4, pp. 899–903, 2010, doi: 10.1111/j.1365-2133.2010.09649.x.10.1111/j.1365-2133.2010.09649.xSearch in Google Scholar PubMed

[22] J. E. Cobb, S. J. White, S. B. Harrap, and J. A. Ellis, “Androgen receptor copy number variation and androgenetic alopecia: a case-control study,” PLoS One, vol. 4, no. 4, p. e5081, 2009, doi: 10.1371/journal.pone.0005081.10.1371/journal.pone.0005081Search in Google Scholar PubMed PubMed Central

[23] J. E. Cobb, S. G. Zaloumis, K. J. Scurrah, S. B. Harrap, and J. A. Ellis, “Evidence for two independent functional variants for androgenetic alopecia around the androgen receptor gene,” Exp Dermatol, vol. 19, no. 11, pp. 1026–1028, 2010, doi: 10.1111/j.1600-0625.2010.01132.x.10.1111/j.1600-0625.2010.01132.xSearch in Google Scholar PubMed

[24] J. A. Ellis, K. J. Scurrah, J. E. Cobb, S. G. Zaloumis, A. E. Duncan, and S. B. Harrap, “Baldness and the androgen receptor: The AR polyglycine repeat polymorphism does not confer susceptibility to androgenetic alopecia,” Hum Genet, vol. 121, no. 3–4, pp. 451–457, May 2007, doi: 10.1007/s00439-006-0317-8.10.1007/s00439-006-0317-8Search in Google Scholar PubMed

[25] J. A. Ellis, M. Stebbing, and S. B. Harrap, “Polymorphism of the androgen receptor gene is associated with male pattern baldness,” J Invest Dermatol, vol. 116, no. 3, pp. 452–455, 2001, doi: 10.1046/j.1523-1747.2001.01261.x.10.1046/j.1523-1747.2001.01261.xSearch in Google Scholar PubMed

[26] V. M. Hayes et al., “The E211 G>A androgen receptor polymorphism is associated with a decreased risk of metastatic prostate cancer and androgenetic alopecia,” Cancer Epidemiology Biomarkers and Prevention, vol. 14, no. 4, pp. 993–996, 2005, doi: 10.1158/1055-9965.EPI-04-0778.10.1158/1055-9965.EPI-04-0778Search in Google Scholar PubMed

[27] A. M. Hillmer et al., “Genetic variation in the human androgen receptor gene is the major determinant of common early-onset androgenetic alopecia,” Am J Hum Genet, vol. 77, no. 1, pp. 140–148, 2005, doi: S0002-9297(07)60910-0 [pii] 10.1086/431425.10.1086/431425Search in Google Scholar PubMed PubMed Central

[28] E. Levy-Nissenbaum, M. Bar-Natan, M. Frydman, and E. Pras, “Confirmation of the association between male pattern baldness and the androgen receptor gene,” Eur J Dermatol, vol. 15, no. 5, pp. 339–340, 2005, doi: 10.1177/120347549800300103.Search in Google Scholar

[29] M. E. Sawaya and A. R. Shalita, “Androgen receptor polymorphisms (CAG repeat lengths) in androgenetic alopecia, hirsutism, and acne,” J Cutan Med Surg, vol. 3, no. 1, pp. 9–15, 1998, doi: 10.1177/120347549800300103.10.1177/120347549800300103Search in Google Scholar PubMed

[30] A. Buniello et al., “The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019,” Nucleic Acids Res, vol. 47, no. D1, pp. D1005–D1012, Jan. 2019, doi: 10.1093/NAR/GKY1120.10.1093/nar/gky1120Search in Google Scholar PubMed PubMed Central

[31] A. M. Hillmer et al., “Susceptibility variants for male-pattern baldness on chromosome 20p11,” Nat Genet, vol. 40, no. 11, pp. 1279–1281, 2008, doi: 10.1038/ng.228.10.1038/ng.228Search in Google Scholar PubMed

[32] J. B. Richards et al., “Male-pattern baldness susceptibility locus at 20p11,” Nat Genet, vol. 40, no. 11, pp. 1282–1284, 2008, doi: 10.1038/ng.255.10.1038/ng.255Search in Google Scholar PubMed PubMed Central

[33] K. Watanabe, E. Taskesen, A. van Bochoven, and D. Posthuma, “Functional mapping and annotation of genetic associations with FUMA.,” Nat Commun, vol. 8, no. 1, p. 1826, 2017, doi: 10.1038/s41467-017-01261-5.10.1038/s41467-017-01261-5Search in Google Scholar PubMed PubMed Central

[34] F. F. Brockschmidt et al., “Susceptibility variants on chromosome 7p21.1 suggest HDAC9 as a new candidate gene for male-pattern baldness,” British Journal of Dermatology, vol. 165, no. 6, pp. 1293–1302, Dec. 2011, doi: 10.1111/J.1365-2133.2011.10708.X.10.1111/j.1365-2133.2011.10708.xSearch in Google Scholar PubMed

[35] R. Li et al., “Six novel susceptibility Loci for early-onset androgenetic alopecia and their unexpected association with common diseases,” PLoS Genet, vol. 8, no. 5, p. e1002746, 2012, doi: 10.1371/journal.pgen.1002746 PGENETICS-D-11-02648 [pii].10.1371/journal.pgen.1002746Search in Google Scholar PubMed PubMed Central

[36] S. Heilmann et al., “Androgenetic Alopecia: Identification of Four Genetic Risk Loci and Evidence for the Contribution of WNT Signaling to Its Etiology,” Journal of Investigative Dermatology, vol. 133, no. 6, pp. 1489–1496, Jun. 2013, doi: 10.1038/JID.2013.43.Search in Google Scholar

[37] S. P. Hagenaars et al., “Genetic prediction of male pattern baldness,” PLoS Genet, vol. 13, no. 2, p. e1006594, Feb. 2017, doi: 10.1371/journal.pgen.1006594.10.1371/journal.pgen.1006594Search in Google Scholar PubMed PubMed Central

[38] S. Heilmann-Heimbach et al., “Meta-analysis identifies novel risk loci and yields systematic insights into the biology of male-pattern baldness.,” Nat Commun, vol. 8, p. 14694, Mar. 2017, doi: 10.1038/ncomms14694.10.1038/ncomms14694Search in Google Scholar PubMed PubMed Central

[39] N. Pirastu et al., “GWAS for male-pattern baldness identifies 71 susceptibility loci explaining 38 % of the risk,” Nat Commun, vol. 8, no. 1, p. 1584, Dec. 2017, doi: 10.1038/s41467-017-01490-8.10.1038/s41467-017-01490-8Search in Google Scholar PubMed PubMed Central

[40] J. K. Pickrell, T. Berisa, J. Z. Liu, L. Segurel, J. Y. Tung, and D. A. Hinds, “Detection and interpretation of shared genetic influences on 42 human traits,” Nat Genet, vol. 48, no. 7, pp. 709–717, 2016, doi: 10.1038/ng.3570.10.1038/ng.3570Search in Google Scholar PubMed PubMed Central

[41] T. Lappalainen, “Functional genomics bridges the gap between quantitative genetics and molecular biology,” Genome Res, vol. 25, no. 10, p. 1427, Oct. 2015, doi: 10.1101/GR.190983.115.10.1101/gr.190983.115Search in Google Scholar PubMed PubMed Central

[42] T. H. Pers et al., “Biological interpretation of genome-wide association studies using predicted gene functions,” Nat Commun, vol. 6, no. 1, pp. 1–9, Jan. 2015, doi: 10.1038/ncomms6890.10.1038/ncomms6890Search in Google Scholar PubMed PubMed Central

[43] S. Heilmann-Heimbach, L. M. Hochfeld, S. K. Henne, and M. M. Nöthen, “Hormonal regulation in male androgenetic alopecia-Sex hormones and beyond: Evidence from recent genetic studies,” Exp Dermatol, vol. 29, no. 9, pp. 814–827, Sep. 2020, doi: 10.1111/EXD.14130.10.1111/exd.14130Search in Google Scholar PubMed

[44] S. Heilmann et al., “Androgenetic alopecia: Identification of four genetic risk loci and evidence for the contribution of WNT signaling to its etiology,” Journal of Investigative Dermatology, vol. 133, no. 6, pp. 1489–1496, Jun. 2013, doi: 10.1038/jid.2013.43.10.1038/jid.2013.43Search in Google Scholar PubMed

[45] L. M. Hochfeld, A. Keller, T. Anhalt, N. Fricker, M. M. Nöthen, and S. Heilmann-Heimbach, “Insights into Male Androgenetic Alopecia: Differential Gene Expression Profiling of Plucked Hair Follicles and Integration with Genetic Data,” Journal of Investigative Dermatology, vol. 139, no. 1, pp. 235–238, Jan. 2019, doi: 10.1016/J.JID.2018.06.182.10.1016/j.jid.2018.06.182Search in Google Scholar PubMed

[46] L. M. Hochfeld et al., “Evidence for a functional interaction of WNT10A and EBF1 in male-pattern baldness,” PLoS One, vol. 16, no. 9, Sep. 2021, doi: 10.1371/JOURNAL.PONE.0256846.10.1371/journal.pone.0256846Search in Google Scholar PubMed PubMed Central

[47] S. Ahouansou, P. le Toumelin, B. Crickx, and V. Descamps, “Association of androgenetic alopecia and hypertension,” European Journal of Dermatology, vol. 17, no. 3, pp. 220–222, May 2007, doi: 10.1684/ejd.2007.0152.Search in Google Scholar

[48] E. S. Ford, D. S. Freedman, and T. Byers, “Baldness and ischemic heart disease in a national sample of men,” Am J Epidemiol, vol. 143, no. 7, pp. 651–657, Apr. 1996, doi: 10.1093/oxfordjournals.aje.a008797.10.1093/oxfordjournals.aje.a008797Search in Google Scholar PubMed

[49] P. A. Lotufo, C. U. Chae, U. A. Ajani, C. H. Hennekens, and J. A. E. Manson, “Male pattern baldness and coronary heart disease: The physicians’ health study,” Arch Intern Med, vol. 160, no. 2, pp. 165–171, Jan. 2000, doi: 10.1001/archinte.160.2.165.10.1001/archinte.160.2.165Search in Google Scholar PubMed

[50] C. R. Herrera, R. B. D’agostino, B. B. Gerstman, L. A. Bosco, and A. J. Belanger, “Baldness and coronary heart disease rates in men from the framingham study,” Am J Epidemiol, vol. 142, no. 8, pp. 828–833, Oct. 1995, doi: 10.1093/oxfordjournals.aje.a117722.10.1093/oxfordjournals.aje.a117722Search in Google Scholar PubMed

[51] S. Lesko, L. Rosenberg, and S. Shapiro, “A Case-Control Study of Baldness in Relation to Myocardial Infarction in Men,” JAMA: The Journal of the American Medical Association, vol. 269, no. 8, pp. 998–1003, Feb. 1993, doi: 10.1001/jama.1993.03500080046030.10.1001/jama.269.8.998Search in Google Scholar

[52] N. Trieu and G. D. Eslick, “Alopecia and its association with coronary heart disease and cardiovascular risk factors: A meta-analysis,” Int J Cardiol, vol. 176, no. 3, pp. 687–695, Oct. 2014, doi: 10.1016/j.ijcard.2014.07.079.10.1016/j.ijcard.2014.07.079Search in Google Scholar PubMed

[53] B. K. Kim, S. J. Choe, H. C. Chung, S. S. Oh, and W. S. Lee, “Gender-specific risk factors for androgenetic alopecia in the Korean general population: Associations with medical comorbidities and general health behaviors,” Int J Dermatol, vol. 57, no. 2, pp. 183–192, Feb. 2018, doi: 10.1111/IJD.13843.10.1111/ijd.13843Search in Google Scholar PubMed

[54] R. Arefi, M. Pishgahi, A. Joharimoghaddam, M. A. Momeni, and M. Khoshfetrat, “Association of Baldness with Coronary Artery Disease and Its Severity,” Galen medical journal, vol. 9, p. e1474, Oct. 2020, doi: 10.31661/GMJ.V9I0.1474.10.31661/gmj.v9i0.1474Search in Google Scholar PubMed PubMed Central

[55] J. G. González-González et al., “Androgenetic alopecia and insulin resistance in young men,” Clin Endocrinol (Oxf), vol. 71, no. 4, pp. 494–499, 2009, doi: 10.1111/j.1365-2265.2008.03508.x.10.1111/j.1365-2265.2008.03508.xSearch in Google Scholar PubMed

[56] P. Hirsso et al., “Association of insulin resistance linked diseases and hair loss in elderly men. Finnish population-based study,” Cent Eur J Public Health, vol. 14, no. 2, pp. 78–81, 2006, doi: 10.21101/cejph.b0045.10.21101/cejph.b0045Search in Google Scholar PubMed

[57] V. Matilainen, P. Koskela, and S. Keinänen-Kiukaanniemi, “Early androgenetic alopecia as a marker of insulin resistance,” Lancet, vol. 356, no. 9236, pp. 1165–1166, Sep. 2000, doi: 10.1016/S0140-6736(00)02763-X.10.1016/S0140-6736(00)02763-XSearch in Google Scholar PubMed

[58] O. A. Bakry, M. A. M. Shoeib, M. K. el Shafiee, and A. Hassan, “Androgenetic alopecia, metabolic syndrome, and insulin resistance: Is there any association? A case–control study,” Indian Dermatol Online J, vol. 5, no. 3, p. 276, 2014, doi: 10.4103/2229-5178.137776.10.4103/2229-5178.137776Search in Google Scholar PubMed PubMed Central

[59] C. C. Yang, F. N. Hsieh, L. Y. Lin, C. K. Hsu, H. M. Sheu, and W. Chen, “Higher body mass index is associated with greater severity of alopecia in men with male-pattern androgenetic alopecia in Taiwan: A cross-sectional study,” J Am Acad Dermatol, vol. 70, no. 2, pp. 297–302.e1, Feb. 2014, doi: 10.1016/j.jaad.2013.09.036.10.1016/j.jaad.2013.09.036Search in Google Scholar PubMed

[60] M. R. Swaroop, B. M. Kumar, B. D. Sathyanarayana, D. Yogesh, J. C. Raghavendra, and P. Kumari, “The Association of Metabolic Syndrome and Insulin Resistance in Early-Onset Androgenetic Alopecia in Males: A Case-Control Study,” Indian J Dermatol, vol. 64, no. 1, pp. 23–27, Jan. 2019, doi: 10.4103/IJD.IJD_724_16.10.4103/ijd.IJD_724_16Search in Google Scholar PubMed PubMed Central

[61] C. Lie, C. F. Liew, and H. H. Oon, “Alopecia and the metabolic syndrome,” Clin Dermatol, vol. 36, no. 1, pp. 54–61, Jan. 2018, doi: 10.1016/J.CLINDERMATOL.2017.09.009.10.1016/j.clindermatol.2017.09.009Search in Google Scholar PubMed

[62] A. M. Hamed, M. A. Fatah, and G. M. Shams, “Androgenetic Alopecia and Metabolic Syndrome: Is Alarin a Missing Link?,” J Clin Aesthet Dermatol, vol. 15, no. 7, p. 32, 2022.Search in Google Scholar

[63] K. Dharam Kumar, Y. Kishan Kumar, and V. Neladimmanahally, “Association of Androgenetic Alopecia with Metabolic Syndrome: A Case-control Study on 100 Patients in a Tertiary Care Hospital in South India,” Indian J Endocrinol Metab, vol. 22, no. 2, p. 196, 2018, doi: 10.4103/IJEM.IJEM_650_17.10.4103/ijem.IJEM_650_17Search in Google Scholar PubMed PubMed Central

[64] S. Arias-Santiago et al., “Androgenetic alopecia as an early marker of benign prostatic hyperplasia,” J Am Acad Dermatol, vol. 66, no. 3, pp. 401–408, Mar. 2012, doi: 10.1016/j.jaad.2010.12.023.10.1016/j.jaad.2010.12.023Search in Google Scholar PubMed

[65] B. R. Oh et al., “Association of benign prostatic hyperplasia with male pattern baldness,” Urology, vol. 51, no. 5, pp. 744–748, May 1998, doi: 10.1016/S0090-4295(98)00108-3.10.1016/S0090-4295(98)00108-3Search in Google Scholar

[66] G. G. Giles et al., “Androgenetic alopecia and prostate cancer: Findings from an Australian case-control study,” Cancer Epidemiology Biomarkers and Prevention, vol. 11, no. 6, pp. 549–553, 2002.Search in Google Scholar

[67] E. Hawk, R. A. Breslow, and B. I. Graubard, “Male pattern baldness and clinical prostate cancer in the epidemiologic follow-up of the first National Health and Nutrition Examination Survey,” Cancer Epidemiology Biomarkers and Prevention, vol. 9, no. 5, pp. 523–527, May 2000.Search in Google Scholar

[68] L. M. O’Keeffe, M. Frysz, J. A. Bell, L. D. Howe, and A. Fraser, “Puberty timing and adiposity change across childhood and adolescence: disentangling cause and consequence,” Human Reproduction, vol. 35, no. 12, pp. 2784–2792, Dec. 2020, doi: 10.1093/HUMREP/DEAA213.10.1093/humrep/deaa213Search in Google Scholar PubMed PubMed Central

[69] D. Horáková et al., “What are the real associations of homeostasis model assessment (HOMA) with body mass index and age?,” Endokrynol Pol, vol. 73, no. 4, pp. 736–742, 2022, doi: 10.5603/EP.A2022.0031.10.5603/EP.a2022.0031Search in Google Scholar PubMed

[70] H. Stefansson et al., “A common inversion under selection in Europeans,” Nat Genet, vol. 37, no. 2, pp. 129–137, Feb. 2005, doi: 10.1038/NG1508.10.1038/ng1508Search in Google Scholar PubMed

[71] A. H. Carey et al., “Polycystic ovaries and premature male pattern baldness are associated with one allele of the steroid metabolism gene CYP17,” Hum Mol Genet, vol. 3, no. 10, pp. 1873–1876, Oct. 1994, doi: 10.1093/HMG/3.10.1873.10.1093/hmg/3.10.1873Search in Google Scholar PubMed

[72] L. Starka, M. Duskova, I. Cermakova, J. Vrbiková, and M. Hill, “Premature androgenic alopecia and insulin resistance. Male equivalent of polycystic ovary syndrome?,” Endocr Regul, vol. 39, no. 4, pp. 127–131, 2005.Search in Google Scholar

[73] F. Day et al., “Large-scale genome-wide meta-analysis of polycystic ovary syndrome suggests shared genetic architecture for different diagnosis criteria,” PLoS Genet, vol. 14, no. 12, Dec. 2018, doi: 10.1371/journal.pgen.1007813.10.1371/journal.pgen.1007813Search in Google Scholar PubMed PubMed Central

[74] E. Fondell, K. C. Fitzgerald, G. J. Falcone, É. J. O’Reilly, and A. Ascherio, “Early-Onset Alopecia and Amyotrophic Lateral Sclerosis: A Cohort Study,” Am J Epidemiol, vol. 178, no. 7, pp. 1146–1149, Oct. 2013, doi: 10.1093/AJE/KWT096.10.1093/aje/kwt096Search in Google Scholar PubMed PubMed Central

[75] H. Moravvej, M. R. Pourani, M. Baghani, and F. Abdollahimajd, “Androgenetic alopecia and COVID‐19: A review of the hypothetical role of androgens,” Dermatol Ther, vol. 34, no. 4, Jul. 2021, doi: 10.1111/DTH.15004.10.1111/dth.15004Search in Google Scholar PubMed PubMed Central

[76] S. K. Henne, L. M. Hochfeld, C. Maj, M. M. Nöthen, and S. Heilmann‐Heimbach, “Systematic investigation of a potential epidemiological and genetic association between male androgenetic alopecia and COVID‐19,” Skin Health and Disease, vol. 1, no. 4, p. e72, Dec. 2021, doi: 10.1002/ski2.72.10.1002/ski2.72Search in Google Scholar PubMed PubMed Central

[77] M. Baghani et al., “Androgenetic alopecia and COVID‐19: Is there a clinical connection?,” J Cosmet Dermatol, vol. 21, no. 2, pp. 420–425, Feb. 2022, doi: 10.1111/jocd.14670.10.1111/jocd.14670Search in Google Scholar PubMed

[78] S. Torabi, M. Mozdourian, R. Rezazadeh, A. Payandeh, S. Badiee, and E. Darchini‐Maragheh, “Androgenetic alopecia in women and men is not related to COVID‐19 infection severity: a prospective cohort study of hospitalized COVID‐19 patients,” Journal of the European Academy of Dermatology and Venereo-logy, vol. 35, no. 9, pp. e553–e556, Sep. 2021, doi: 10.1111/jdv.17353.10.1111/jdv.17353Search in Google Scholar PubMed PubMed Central

[79] H. M. Tanha, S. Medland, N. G. Martin, and D. R. Nyholt, “Genetic correlation analysis does not associate male pattern baldness with COVID-19,” J Am Acad Dermatol, vol. 85, no. 4, pp. 971–973, Oct. 2021, doi: 10.1016/j.jaad.2021.05.009.10.1016/j.jaad.2021.05.009Search in Google Scholar PubMed PubMed Central

[80] H. Blasco et al., “Amyotrophic lateral sclerosis: a hormonal condition?,” Amyotroph Lateral Scler, vol. 13, no. 6, pp. 585–588, Oct. 2012, doi: 10.3109/17482968.2012.706303.10.3109/17482968.2012.706303Search in Google Scholar PubMed

[81] P. Wicks, “Hypothesis: higher prenatal testosterone predisposes ALS patients to improved athletic performance and manual professions,” Amyotroph Lateral Scler, vol. 13, no. 3, pp. 251–253, May 2012, doi: 10.3109/17482968.2011.634009.10.3109/17482968.2011.634009Search in Google Scholar PubMed PubMed Central

[82] R. L. Rosenfield and D. A. Ehrmann, “The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited,” Endocr Rev, vol. 37, no. 5, p. 467, 2016, doi: 10.1210/ER.2015-1104.10.1210/er.2015-1104Search in Google Scholar PubMed PubMed Central

[83] P. Szulc, “Role of sex steroids hormones in the regulation of bone metabolism in men: Evidence from clinical studies,” Best Practice and Research: Clinical Endocrinology and Metabolism, vol. 36, no. 2. Bailliere Tindall Ltd, Mar. 01, 2022. doi: 10.1016/j.beem.2022.101624.10.1016/j.beem.2022.101624Search in Google Scholar PubMed

[84] G. Corona et al., “Testosterone supplementation and bone parameters: a systematic review and meta-analysis study,” Journal of Endocrinological Investigation, vol. 45, no. 5. Springer Science and Business Media Deutschland GmbH, pp. 911–926, May 01, 2022. doi: 10.1007/s40618-021-01702-5.10.1007/s40618-021-01702-5Search in Google Scholar PubMed

[85] M. Marcińska et al., “Evaluation of DNA variants associated with androgenetic alopecia and their potential to predict male pattern baldness,” PLoS One, vol. 10, no. 5, May 2015, doi: 10.1371/journal.pone.0127852.10.1371/journal.pone.0127852Search in Google Scholar PubMed PubMed Central

[86] F. Liu et al., “Prediction of male-pattern baldness from genotypes,” European Journal of Human Genetics, vol. 24, no. 6, pp. 895–902, Jun. 2016, doi: 10.1038/ejhg.2015.220.10.1038/ejhg.2015.220Search in Google Scholar PubMed PubMed Central

[87] Y. Chen et al., “Genetic prediction of male pattern baldness based on large independent datasets,” Eur J Hum Genet, 2022, doi: 10.1038/S41431-022-01201-Y.10.1038/s41431-022-01201-ySearch in Google Scholar PubMed PubMed Central

[88] A. R. Martin, M. Kanai, Y. Kamatani, Y. Okada, B. M. Neale, and M. J. Daly, “Clinical use of current polygenic risk scores may exacerbate health disparities,” Nat Genet, vol. 51, no. 4, pp. 584–591, Apr. 2019, doi: 10.1038/S41588-019-0379-X.10.1038/s41588-019-0379-xSearch in Google Scholar PubMed PubMed Central

[89] R. H. True, “Is Every Patient of Hair Loss a Candidate for Hair Transplant?-Deciding Surgical Candidacy in Pattern Hair Loss,” Indian Journal of Plastic Surgery, vol. 54, no. 4. Georg Thieme Verlag, pp. 435–440, Oct. 01, 2021. doi: 10.1055/s-0041-1739247.10.1055/s-0041-1739247Search in Google Scholar PubMed PubMed Central

[90] F. Xu et al., “Prevalence and types of androgenetic alopecia in Shanghai, China: a community-based study,” British Journal of Dermatology, vol. 160, no. 3, pp. 629–632, Mar. 2009, doi: 10.1111/j.1365-2133.2008.08909.x.10.1111/j.1365-2133.2008.08909.xSearch in Google Scholar PubMed

[91] J.-H. Paik, J.-B. Yoon, W.-Y. Sim, B.-S. Kim, and N.-I. Kim, “The prevalence and types of androgenetic alopecia in Korean men and women,” British Journal of Dermatology, vol. 145, no. 1, pp. 95–99, Jul. 2001, doi: 10.1046/j.1365-2133.2001.04289.x.10.1046/j.1365-2133.2001.04289.xSearch in Google Scholar PubMed

[92] L. R. Setty, “Hair patterns of the scalp of white and Negro males,” Am J Phys Anthropol, vol. 33, no. 1, pp. 49–55, 1970, doi: 10.1002/ajpa.1330330108.10.1002/ajpa.1330330108Search in Google Scholar PubMed

[93] K. Adhikari et al., “A genome-wide association scan in admixed Latin Americans identifies loci influencing facial and scalp hair features,” Nat Commun, vol. 7, Mar. 2016, doi: 10.1038/ncomms10815.10.1038/ncomms10815Search in Google Scholar PubMed PubMed Central

[94] B. Liang et al., “Genetic Variants at 20p11 Confer Risk to Androgenetic Alopecia in the Chinese Han Population,” PLoS One, vol. 8, no. 8, Aug. 2013, doi: 10.1371/JOURNAL.PONE.0071771.10.1371/journal.pone.0071771Search in Google Scholar PubMed PubMed Central

[95] I. Y. Kim et al., “The first broad replication study of SNPs and a pilot genome-wide association study for androgenetic alopecia in Asian populations,” J Cosmet Dermatol, 2022, doi: 10.1111/JOCD.15187.10.1111/jocd.15187Search in Google Scholar PubMed

[96] P. M. Ramos and H. A. Miot, “Female pattern hair loss: A clinical and pathophysiological review,” Anais Brasileiros de Dermatologia, vol. 90, no. 4. Sociedade Brasileira de Dermatologia, pp. 529–543, Jul. 01, 2015. doi: 10.1590/abd1806-4841.20153370.10.1590/abd1806-4841.20153370Search in Google Scholar PubMed PubMed Central

[97] S. Redler, A. G. Messenger, and R. C. Betz, “Genetics and other factors in the aetiology of female pattern hair loss,” Exp Dermatol, vol. 26, no. 6, pp. 510–517, Jun. 2017, doi: 10.1111/exd.13373.10.1111/exd.13373Search in Google Scholar PubMed

[98] A. Łukasik, K. Kozicka, A. Kłosowicz, A. Jaworek, and A. Wojas-Pelc, “The role of family history and its influence on the onset time in female pattern hair loss,” Postepy Dermatol Alergol, vol. 38, no. 5, pp. 815–818, 2021, doi: 10.5114/ada.2020.100745.10.5114/ada.2020.100745Search in Google Scholar PubMed PubMed Central

[99] M. A. Smith and R. S. Wells, “Male-Type Alopecia, Alopecia Areata, and Normal Hair in Women: Family Histories,” Arch Dermatol, vol. 89, no. 1, pp. 95–98, Jan. 1964, doi: 10.1001/archderm.1964.01590250101017.10.1001/archderm.1964.01590250101017Search in Google Scholar PubMed

[100] L. Yip et al., “Gene-wide association study between the aromatase gene (CYP19A1) and female pattern hair loss,” Br J Dermatol, vol. 161, no. 2, pp. 289–294, 2009, doi: 10.1111/J.1365-2133.2009.09186.X.10.1111/j.1365-2133.2009.09186.xSearch in Google Scholar PubMed

[101] A. Łukasik, K. Kozicka, A. Pisarek, and A. Wojas-Pelc, “The role of CYP19A1 and ESR2 gene polymorphisms in female androgenetic alopecia in the Polish population,” Advances in Dermatology and Allergology/Postȩpy Dermatologii i Alergologii, vol. 39, no. 4, p. 708, 2022, doi: 10.5114/ADA.2021.108429.10.5114/ada.2021.108429Search in Google Scholar PubMed PubMed Central

[102] S. Redler et al., “Investigation of variants of the aromatase gene (CYP19A1) in female pattern hair loss,” British Journal of Dermatology, vol. 165, no. 3, pp. 703–705, Sep. 2011, doi: 10.1111/J.1365-2133.2011.10456.X.10.1111/j.1365-2133.2011.10456.xSearch in Google Scholar PubMed

[103] W. Rui et al., “Polymorphic CAG Repeat Numbers in the Androgen Receptor Gene of Female Pattern Hair Loss in a Han Chinese Population,” Dermatology, vol. 232, no. 4, pp. 464–467, Sep. 2016, doi: 10.1159/000446648.10.1159/000446648Search in Google Scholar PubMed

[104] W. Rui et al., “Association of Single Nucleotide Polymorphisms in the CYP19A1 Gene with Female Pattern Hair Loss in a Chinese Population,” Dermatology, vol. 231, no. 3, pp. 239–244, Dec. 2015, doi: 10.1159/000433597.10.1159/000433597Search in Google Scholar PubMed

[105] J. Ohn et al., “Early onset female pattern hair loss: A case-control study for analyzing clinical features and genetic variants,” J Dermatol Sci, vol. 106, no. 1, pp. 21–28, Apr. 2022, doi: 10.1016/J.JDERMSCI.2022.02.011.10.1016/j.jdermsci.2022.02.011Search in Google Scholar PubMed

[106] S. Redler et al., “The oestrogen receptor 2 (ESR2) gene in female-pattern hair loss: replication of association with rs10137185 in German patients,” British Journal of Dermatology, vol. 170, no. 4, pp. 982–985, Apr. 2014, doi: 10.1111/BJD.12756.10.1111/bjd.12756Search in Google Scholar PubMed

[107] B. Liang, Y. Ding, Y. Zhou, C. Yang, and Z. Cheng, “Evaluation of susceptibility genes/loci associated with male androgenetic alopecia (maga) for female-pattern hair loss in a chinese han population and a brief literature review,” Medical Science Monitor, vol. 27. International Scientific Information, Inc., pp. e933424-1, 2021. doi: 10.12659/MSM.933424.10.12659/MSM.933424Search in Google Scholar PubMed PubMed Central

[108] S. Redler et al., “Selected variants of the steroid-5-alpha-reductase isoforms SRD5A1 and SRD5A2 and the sex steroid hormone receptors ESR1, ESR2 and PGR: No association with female pattern hair loss identified,” Exp Dermatol, vol. 21, no. 5, pp. 390–393, May 2012, doi: 10.1111/J.1600-0625.2012.01469.X.10.1111/j.1600-0625.2012.01469.xSearch in Google Scholar PubMed

[109] S. Redler et al., “Investigation of the male pattern baldness major genetic susceptibility loci AR/EDA2R and 20p11 in female pattern hair loss,” British Journal of Dermatology, vol. 166, no. 6, pp. 1314–1318, Jun. 2012, doi: 10.1111/J.1365-2133.2012.10877.X.10.1111/j.1365-2133.2012.10877.xSearch in Google Scholar PubMed

[110] S. Redler et al., “Investigation of six novel susceptibility loci for male androgenetic alopecia in women with female pattern hair loss,” J Dermatol Sci, vol. 72, no. 2, pp. 186–188, Nov. 2013, doi: 10.1016/J.JDERMSCI.2013.06.012.10.1016/j.jdermsci.2013.06.012Search in Google Scholar PubMed

[111] R. Nuwaihyd et al., “Investigation of four novel male androgenetic alopecia susceptibility loci: No association with female pattern hair loss,” Arch Dermatol Res, vol. 306, no. 4, pp. 413–418, Dec. 2014, doi: 10.1007/S00403-013-1436-4/TABLES/2.10.1007/s00403-013-1436-4Search in Google Scholar PubMed

[112] J. D. Backman et al., “Exome sequencing and analysis of 454,787 UK Biobank participants,” Nature 2021 599:7886, vol. 599, no. 7886, pp. 628–634, Oct. 2021, doi: 10.1038/s41586-021-04103-z.10.1038/s41586-021-04103-zSearch in Google Scholar PubMed PubMed Central

[113] Q. Wang et al., “Rare variant contribution to human disease in 281,104 UK Biobank exomes,” Nature 2021 597:7877, vol. 597, no. 7877, pp. 527–532, Aug. 2021, doi: 10.1038/s41586-021-03855-y.10.1038/s41586-021-03855-ySearch in Google Scholar PubMed PubMed Central

[114] I. Dunham et al., “An Integrated Encyclopedia of DNA Elements in the Human Genome,” Nature, vol. 489, no. 7414, p. 57, Sep. 2012, doi: 10.1038/NATURE11247.10.1038/nature11247Search in Google Scholar PubMed PubMed Central

[115] M. R. Nelson et al., “The support of human genetic evidence for approved drug indications,” Nature Genetics 2015 47:8, vol. 47, no. 8, pp. 856–860, Jun. 2015, doi: 10.1038/ng.3314.10.1038/ng.3314Search in Google Scholar PubMed

[116] E. A. King, J. Wade Davis, and J. F. Degner, “Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval,” PLoS Genet, vol. 15, no. 12, p. e1008489, 2019, doi: 10.1371/JOURNAL.PGEN.1008489.10.1371/journal.pgen.1008489Search in Google Scholar PubMed PubMed Central

Published Online: 2023-04-05
Published in Print: 2023-04-03

© 2023 bei den Autorinnen und Autoren, publiziert von De Gruyter.

Dieses Werk ist lizensiert unter einer Creative Commons Namensnennung 4.0 International Lizenz.

Downloaded on 2.6.2024 from https://www.degruyter.com/document/doi/10.1515/medgen-2023-2003/html
Scroll to top button