Register Now for NAAF's 39th Annual Patient Conference June 27-30, 2024 in Washington, D.C.!

Funded Research

Laboratory concept; Scientist uses a dropper to transfer chemical reagent to test tube. He observes the chemical reaction with a blurred background of research in the laboratory

NAAF’s research program funds investigations that advance the understanding of the mechanisms behind alopecia areata, help identify new therapeutic targets, provide information on the biological, psychosocial, and economic impacts, and further knowledge toward treatments and a cure. Since 1985, NAAF has awarded more than 208 research grants totaling more than $5.7 million. The studies we’ve invested in have enabled significant advances in research by helping to unravel the genetics of alopecia areata, identify important immune pathways, and yield clues to targeted treatments.

Find out about applying for a grant on the Research Grants and Awards page.

Use of microneedles for the management of alopecia areata

Year: 2024 PI Name: Natalie Artzi, PhD; Jamil R. Azzi, MD, PhD Award Type: Research Grant Status: In Progress Summary:
Alopecia Areata (AA) is a skin condition where patients experience hair loss due to a dysregulation of the immune system. The only solution to date requires local injection of corticosteroids, which is painful, or the use of immunosuppressants, which cause severe side effects and are associated with high rates of relapse as the hair falls again after treatment. Our group has developed a revolutionary approach to treat this disease by using a novel medical device containing tiny, microscopic needles—a microneedle patch. Given the dimensions of the microneedles, less than a millimeter in length, the patch can be self-administered and is painless. This is particularly important for kids and for patients where disease is manifested in areas that cannot be injected like the eyebrows. The patch can release drug molecules through the skin directly into the bald lesions to restore the balance in the immune system and encourage hair growth.

Read more

Abstract:
Over the past two years, our multidisciplinary team including clinicians and biomedical engineers has focused on the development of revolutionary microneedle platform for the management of AA. In this proposal, we aim to rectify the local Treg deficiency in AA patients, by recruiting and amplifying Treg function, locally at the affected skin lesion, using our novel hydrogel-based microneedle (MN) platform for chemokine/cytokine delivery. Our proposal is built on the foundation of our work published recently (Artzi, Azzi, and colleagues, Advanced Functional Materials, 2021) 12, where we demonstrated that our engineered polymeric microneedle platform supports local delivery of CCL22 and IL-2 to promote Treg homing and expansion for prevention of allograft rejection in murine skin allograft model. The skin offers unique clinical opportunity for direct and easy access to an intricate network of immune cells including Tregs, which we leveraged for targeted transcutaneous-delivery of immunotherapies using MNs. Our MN-platform fabricated with hyaluronic acid (HA), allowed transdermal delivery of immunomodulators for local action in the skin allografts, while obviating systemic side effects in peripheral organs such as the spleen—a commonly observed phenomenon following systemic delivery of immune-suppressive agents. Moreover, our MNs allow sampling of interstitial skin fluid (ISF), a unique biofluid offering diagnostic insights into the perfusion of analytes. The matrix of our MN platform has been engineered to contain disulfide bonds, enabling us to dissolve the needles following retri eval and isolate cellular biomarkers which can be used to report on the disease state. Based on the foundation of this work and its relevance to restoring Treg homeostasis, we have used our MN platform to deliver chemokines and cytokines locally at the AA lesion site to recruit and restore Treg number and function. In an AA murine model, we have demonstrated that our MNs delivering CCL22 (for Treg recruitment), and IL-2 (for Treg proliferation) remodeled the immune profile at the hair follicle level, increasing Treg frequencies and reversing AA symptoms as targeted, sustained hair regrowth was observed in the AA lesions (manuscript submitted). We also demonstrated that the T cell immune profile in ISF when extracted with the MNs correlate with that in the skin. Here, we aim to expand our understanding on the cellular mechanism driving disease recovery and the potential of Treg-centric therapies, as opposed to immunosuppressive agents, for the management of AA. We predict that our technology solution will be able to provide efficacy and durability of the therapeutic effects, due to the unique mechanism of action of our agent combination that addresses the root cause of AA pathogenesis at the local lesion level, and will harbor the required safety profile by virtue of the minimally invasive, painless topical application of our novel polymeric MN-based platform to deliver nanogram level drug doses, eliminating systemic side effects and local pain. By establishing an optimal balance of the benefit/risk profile, a crucial piece missing from the current treatment strategies, in addition to enhancing the quality of life and compliance for the patient, our technology has the potential to be not only a strong contender, but a game -changer in the AA treatment landscape.

Read more

Impact: Using a miconeedle patch, we have demonstrated in a mouse model that the hair can grow back in the affected areas while avoiding side effects and toxicities given the local and targeted nature of the patch. We believe that our platform will empower AA patients with a safer, superior, and easy-to-use treatment solution.

Identification of autoantigen epitopes in alopecia areata by T-Scan

Year: 2024 PI Name: Zhenpeng Dai, PhD Award Type: Research Grant Status: In Progress Summary:
Alopecia areata (AA) is an autoimmune disease that causes hair loss. Immune cells known as T cells recognize hair follicles’ own components (antigen) as foreign and elicit an immune response against hair follicle. We recently identified several alopecic T cell clones in the mouse model of AA. However, the protein that the alopecic T cells recognize in hair follicle remains unknown. T-Scan is a high-throughput method for genomewide identification of antigens recognized by T cells. To figure out what the T cells are recognizing in AA hair follicles, we will use T-Scan to find own protein components recognized by alopecic T cells.

Read more

Abstract:
Alopecia Areata (AA) is one of the most common autoimmune diseases in the US with a lifetime risk of 1.7%. AA causes significant disfigurement and psychological distress to AAaffected individuals. The etiology of AA is still incompletely understood. The T -cell-mediated autoimmune hypothesis with the collapse of hair follicle (HF) immune privilege is most likely and develops under a genetic background [3]. We recently identified CD8+NKG2D+ T cells as the key pathogenic cells in AA [4]. The role of CD8+NKG2D+ T cells in the process of HF destruction has remained enigmatic. We previously used next-generation sequencing of the TCRβ repertoire in the C3H/HeJ mouse model of AA. We observed interindividual sharing of TCRβ chain protein sequences, which strongly supports a model of antigenic drive in AA. However, the relationship between CD8+ T cell clonality and pathogenicity is poorly understood. We leveraged recent advances in single-cell technology and performed parallel single-cell RNA and T cell receptor (TCR) sequencing in the graft-induced C3H/HeJ mouse model of AA to interrogate time-dependent changes in the AA immune landscape with respect to both gene expression profiles and T cell clonality. Using the highly expanded CD8+ TCR sequences identified in our study, we performed TCR retrogenic mice and TCR engineered T cells to demonstrate that expanded CD8+ T cell clones are sufficient and necessary for AA, establishing the causal relationship between CD8+ T cell clonality and pathogenicity in disease. However, in AA, the antigen epitopes recognized by alopecic T cells has remained unknown, mainly due to the lack of genome-scale and high-throughput antigen discovery tools. Previous studies showed that using online matrix-assisted algorithms could predict MHC binding peptide sequences for autoimmune diseases. One limitation of this method is that only a small number of antigens can be discovered. Cell-based screening methods for CD8+ T cells, such as T-Scan, have achieved proteome-wide success in identifying cognate antigens for TCRs from CD8+ cells. T-Scan utilizes the granzyme-mediated cytolytic ability of CD8+ cells toward target cells that express a library of candidate antigens that are processed and presented endogenously on MHC-I molecules as a functional readout. Given the power of T-Scan in finding T cell epitopes, we postulate that T-Scan would facilitate the discovery of HF auto-antigens recognized by alopecic CD8+ T cells. Successful identification of antigen epitopes in the AA mouse model will provide knowledge to find antigen epitopes in human AA and will enable the development of specific therapeutic modalities targeting antigen - specific T cells.

Read more

Impact: Discovery of specific antigens recognized by alopecic T cells will help us to unravel the mechanisms underlying AA development as well as discovery of new therapeutic avenues

The genetics of alopecia areata: Integration of population studies and clinical data

Year: 2024 PI Name: Michal Linial, PhD Award Type: Research Grant Status: In Progress Summary:
Alopecia Areata (AA) is a rare autoimmune disorder that causes hair loss, typically in small, round patches on the scalp. While the exact cause of AA is not fully understood, genetics is believed to play a significant role in its development. AA tends to run in families, with a higher risk within family members with an affected individual. This research goal is to advance the genetic basis by focusing on the genetic signatures that dictate the AA risk score for individuals. We intend to address the genetics of AA by benefiting from the whole genome and full exomes from the UK Biobank (UKB). In the UKB there are approximately 800 participants with clinical manifestations of AA. With the detailed genome, we intend to: (i) identify significant candidate genes; (ii) seek rare variants that contribute to AA. (ii) Creating a polygenic risk score that focuses on the Chr. 6 MHC-extended loci. (iii) identifying sex-dependent genetic signals; (iv) using time-sensitive comorbidity analysis for the risk hazard for patient stratification. The project aims to compile variants, identify associated genes, infer functionality, and examine sex and age-dependent comorbidity.

Read more

Abstract:
Alopecia areata (AA) is an autoimmune disorder characterized by hair loss in round patches on the scalp. The severity and extent of hair loss vary widely among individuals, and environmental factors (stress episodes, vitamin supply, diet) may affect the severity and flare of the disease. Emotional distress and a reduced quality of life are common consequences. AA is affecting approximately 1:500 to 1:1000 of the global population (McDonagh and Tazi-Ahnini, 2002). AA has a more severe manifestation with complete hair loss that is rare and will not be further discussed. Importantly, AA is not limited to a specific age group, gender, or ethnic background. It often begins in childhood or early adulthood, with females being more susceptible to the severe forms of the condition (Betz et al., 2015; Duvic et al., 2001; Green and Sinclair, 2000). The cause of AA is when immune cells (like T cells) mistakenly attack hair follicles. Like most autoimmune diseases, family history of AA is a risk factor that suggests a genetic component in the development of AA. Individuals with AA may have an increased risk of developing other autoimmune conditions, including thyroid disorders, vitiligo, and atopic dermatitis. Still, the exact triggers and mechanisms behind the dysregulation of the immune response are an enigma (Petukhova et al., 2011; Silverberg, 2022; Sundberg and King, 2003). This research project aims to investigate the genetic factors associated with AA by utilizing stateof-the-art gene-based approaches in the UK biobank dataset (UKB) (Karczewski et al., 2022). The study focuses on identifying genetic signals specific to AA diagnoses and the 5- to 10-fold larger cohort from self-reported data. By analyzing the genetic data of European-origin participants, we aim to identify candidate genes that are significantly associated with AA and explore their potential implications in disease management. One unique aspect of this research is the emphasis on coding genes, which, although they make up only a small percentage of the genome, offer greater interpretability in terms of gene function, pathways, biological relevance, and potential therapeutic targets. By focusing on coding genes, and specifically on the HLA loci, the study aims to provide clearer insights into the genetic mechanisms underlying AA. Comorbidity and gender differences will also be considered in the analysis. By examining the genetic data in relation to comorbidities and considering gender-specific effects, we aim to gain a more comprehensive understanding of the genetic underpinnings of AA. The study will employ complementary association tests in all GWAS, coding-GWAS, SKAT, PWAS, and knowledge-based focused signaling domains (Brandes et al., 2020; Zucker et al., 2023a; Zucker et al., 2023b). These tests will empower the genetic investigation of AA by identifying associated variants and genes. Additionally, the research project aims to infer functional implications from the identified genes and examine age-dependent comorbidities associated with AA. Our preliminary analyses already highlight the power of complementary approaches that benefit from clinical knowledge (Zucker et al., 2023a; Zucker et al., 2023b). The expected outcome of this research is to enhance our understanding of the genetic factors contributing to AA. By identifying candidate genes and potential therapeutic targets, the study aims to improve disease management and the well-being of individuals. The project also incorporates novel aspects, of focusing on the overlooked recessive inheritance and using complementary methods to extract biological relevance. Overall, this research project combines advanced genetic analysis techniques, extensive data resources, and a focus on clinical-genetics applications to shed light on the genetic basis of AA and provide insights that can lead to improved treatments, management strategies for individuals with this condition, and suggested targets for focused therapy.

Read more

Impact: The novelty in our research stems from the integrative approach that focuses on coding gene knowledge, addressing the overlooked recessive inheritance in AA genetics, and using knowledge-based methods to provide a fresh view of this chronic disease that impacts individuals' quality of life from first occurrence through life. The research aims to enhance understanding, identify therapeutic targets, and improve disease management for the benefit of AA patients.

High dimensional characterization of the systemic immune profile of pediatric alopecia areata

Year: 2023 PI Name: Benjamin Ungar, MD; Daniel Lozano-Ojalvo, PhD Award Type: Pediatric AA Challenge Grant Status: In Progress Summary:
Alopecia areata (AA) is a common cause of non-scarring hair loss that affects around 2% of people worldwide. AA is known to be generally caused by the immune system attacking hair follicles, but many details of this process remain unknown. A few studies have suggested that the inflammation in AA is not restricted to the skin but involves systemic immune responses with measurable inflammatory markers in peripheral blood. However, the number of studies is limited and they have just evaluated AA adult patients. The objective of our proposal is to characterize the immune cell profile and circulating inflammatory biomarkers in blood from pediatric AA patients.

Read more

Abstract:
Alopecia areata (AA) is an immune cell-mediated disease characterized by non-scarring hair loss with a 2.1% lifetime risk. The pathogenesis of AA remains incompletely understood, but a key role for T cells damaging the hair follicle is well-established. Recent studies have also demonstrated that AA is not skin-limited; AA is characterized by activated CD4+ and CD8+ T cell subsets in peripheral blood, with corresponding inflammatory biomarkers substantially elevated in serum samples. However, this evidence is restricted to T cell responses in adult AA patients without characterization of other immune cell components and data in pediatric AA subjects are lacking. Our goal, therefore, is to high-dimensionally describe systemic immune pathways involved in pediatric AA by characterizing peripheral innate and adaptive immune populations and their associated biomarkers. We plan to collect blood samples from pediatric subjects (12-18 years) with AA (n=30) and healthy controls (n=30). Phenotypical and functional characteristics of circulating immune cells will be established by spectral flow cytometry (~60 markers). Serum samples will be analyzed using Olink platform for the analysis of ~500 analytes. Circulating biomarkers will be evaluated and correlated with immune cell subsets and clinical severity (Severity of Alopecia Tool, SALT, scores). This proposal will expand our mechanistic understanding of pediatric AA, identifying potential therapeutic targets and paving the way for disease-modifying treatments.

Read more

Impact: Improved understanding of the nuances of the systemic inflammatory process in alopecia areata is important to inform how current treatments should be applied as well as to identify new potential treatments.

Alopecia & Us: Studying the impact of alopecia areata to adolescents and the family

Year: 2023 PI Name: Chantal Cotter, MD Award Type: Pediatric AA Challenge Grant (cofunded with PeDRA) Status: In Progress Summary:
Alopecia areata (AA) is one of the most common hair loss disorders characterized by loss of hair in round patches, usually on the scalp. The exact cause of AA is unknown, but it seems to be caused by the immune system attacking the hair follicles by mistake. Having AA can have impact on a child’s sense of well-being and increase their risk of mental health disorders including depression, anxiety, and OCD. Further, there may an impact on the family’s quality of life. Understanding this burden is paramount to ensuring we understand the full effect AA has on a child. We will do this via an online questionnaire which patients (aged 12-18) and their families can complete at home, anytime, anywhere in the world and in any language. Only through awareness of the full impact of this disease we will be able to advocate for access to treatments but also campaign for recognition and support of mental health in this group.

Read more

Abstract:
Patients with alopecia areata (AA) in the adolescent population have consistently demonstrated poor health related quality of life measures with disease severity and increasing age correlating with poorer quality-of-life scores. In addition, there is research to suggest that there is a high prevalence of psychosocial comorbidities in adolescent patients suffering from AA. We hope to establish a research platform for adolescent patients with AA allowing the phenotypic and in-depth, crosssectional epidemiological studying of the illness. We will also study mental wellbeing, including psychosocial comorbidities, disease perception and stigma. Further we will examine the effect of childhood AA on the family and capture the sequalae of disease on family life. We aim to recruit 1000 patients worldwide in this observational study. Adolescents and their parents will have the opportunity to participate through a self-reporting online platform. Patients will complete several concise e-questionnaire-based metrics, including the Children’s Dermatology Life Quality Index, the Pediatric Index of Emotional Distress, the Family Reported Outcome Measure, the Family Dermatology Life Quality Index and EQ-5DY. This study will enable future research into therapeutic responsiveness by linking treatment effectiveness with research into predictive parameters such as the psychological and socioeconomic status of the adolescent patient and family. Further we hope to highlight the importance of the psychological impact of AA in adolescents and their families prompting a more holistic approach of such patients.

Read more

IL-15 as a Candidate Key Cytokine in Alopecia Areata Pathobiology

Year: 2020 PI Name: Ralf Paus, MD Award Type: Research Grant Status: Completed Summary:
The purpose of this project is to test the hypothesis that blockade of interleukin-15 (IL-15) or IL-15 receptor-alpha can provide therapeutic benefit in alopecia areata.

Read more

Abstract:
Effective alopecia areata (AA) treatment requires selective targeting of the key pathomechanisms to combine optimal, long-lasting therapy outcomes with reduced adverse effects. Recent research has suggested that the multi-function cytokine, interleukin-15 (IL-15), is important in AA pathobiology. Yet, convincing evidence for this remains to be generated in the human system. Specifically, since collapse of the hair follicle’s (HF) physiological immune privilege (IP) is an essential prerequisite for AA lesions to develop while HF-IP restoration is required for hair regrowth, it is critical to clarify the impact of IL-15 on human HF-IP - either directly, or indirectly. Here, we propose to do this by asking (in optimally suited, perfectly complementary human ex vivo and in vivo assay systems, i.e. AA patient skin biopsies, scalp HF organ culture and the humanized AA mouse model) whether: a) IL-15 and/or IL-15 receptor-alpha expression in/around lesional and non-lesional AA HFs is abnormal compared to healthy human scalp skin; b) IL-15 alone induces and/or enhances IFNγ-induced human HF-IP collapse; c) selectively antagonizing IL-15 receptor-alpha mediated signaling prevents experimentally induced HF-IP collapse and/or even restores HF-IP and promotes human hair regrowth in vivo; d) IL-15 promotes IFNγ secretion by pathogenic T/NK cells and/or promotes pathological perifollicular mast cell/T cell interactions in vivo. These clinically highly relevant studies will clarify whether IL-15 signaling really deserves selective targeting in future AA management, which adverse HF effects this could exert, and whether there is subgroup of AA patients who likely will benefit most from this.

Read more

Impact: If successful, this study could provide new insights into the mechanism of alopecia areata pathogenesis mediated by IL-15 with important implications for future therapeutic intervention.

Collective Mechanism of Hair Regrowth during Alopecia Areata Resolution

Year: 2020 PI Name: Maksim Plikus, PhD Award Type: Pilot & Feasibility Grant Status: Completed Summary:
This project aims to evaluate evidence that progression from telogen to anagen growth follows a wave pattern by repeated evaluation with optical coherence tomography imaging; and to perform single cell sequencing to identify factors that predict progression to anagen.

Read more

Abstract:
In alopecia areata (AA), thousands of actively growing (anagen) scalp hair follicles transition into a dormant (telogen) state. When AA resolves, either spontaneously or in response to JAK inhibitor treatment, the newly formed anagen follicles re-emerge rapidly in what grossly appear to be spreading waves. This growth pattern is commonly seen in rodents, where spreading waves are the outcome of a collective behavior, when anagen follicles signal to activate neighboring telogen follicles to also enter anagen in a chain-like reaction. This mechanism leads to a highly efficient anagen activation across large regions of skin. Inspired by our preliminary data on hair regrowth patterns in JAK inhibitor-treated AA patients, in this pilot application we propose to definitively establish the existence of a wave-like mechanism for anagen activation in human scalp, using optical coherence tomography imaging coupled with histological analysis of re-growing wave-front in AA. We will also collect single-cell RNA-sequencing data from wave-front hair-bearing skin and compare its cellular composition and key molecular changes to those of adjacent alopecic areas. High throughput single-cell data will be used to develop hypotheses on cellular and signaling changes that accompany collective AA hair regrowth, previously undescribed in human hair. We will be able to identify hair regeneration-associated changes in intra- and peri-follicular cell types, including immune cells, and key inhibitory and activating signaling factor changes. The hypothesis proposed herein is novel for the AA field, and its results will significantly advance the understanding of AA resolution mechanisms and open new lines of AA research.

Read more

Impact: If successful, this study could provide important mechanistic insights into hair biology, identify novel molecular targets for anagen activation, and lead to the development of new growth factor-based strategies for enhanced hair regeneration in patients with alopecia areata.

Nutraceutical Stimulation of Intrafollicular Autophagy as an Adjuvant Strategy in the Management of Alopecia Areata

Year: 2019 PI Name: Ralf Paus, MD Award Type: Pilot & Feasibility Grant Status: Completed Summary:
This project will explore whether alopecia areata hair follicles display malfunctions in autophagy, which may make them more susceptible to immune-mediated hair follicle damage, and whether promoting hair follicle autophagy with nutraceuticals enhances human hair follicle growth and stress-resistance.

Read more

Abstract:
Although novel treatments for alopecia areata (AA) such as JAK inhibitors can be efficacious, they are extremely expensive, may have long-term adverse effects, and don’t prevent AA relapse after therapy is discontinued. Therefore, new supportive AA therapies remain to be developed that also aid more permanent hair regrowth. Autophagy is a organelle and protein recycling process used by cells to facilitate growth and survival during stress. Recently, we have demonstrated that human scalp hair follicles (HFs) recruit autophagy to sustain their growth, while blocking HF autophagy inhibits human hair growth, Therefore, we propose to explore whether AA HFs display malfunctions in autophagy, which may make them more susceptible to immune-mediated HF damage, and whether promoting HF autophagy with nutraceuticals enhances human HF growth and stress-resistance. Specifically, we will investigate whether the expression of autophagy marker proteins and autophagolysosomes differ between healthy, non-lesional and lesional AA HFs. Next, we study whether interferon- the key cytokine in AA pathogenesis, modulates autophagy (i.e. LC3B and SQSTM1 expression and autophagolysosome generation) and whether inhibiting autophagy by LC3B gene-silencing prevents or promotes interferon-induced damage in organ-cultured human HFs. Finally, we will investigate whether enhancing autophagy by recognized enhancers of autophagic flux, i.e. the nutritional supplements caffeine and/or methylspermidine, makes cultured human HFs more resistant to interferon-induced HF damage and growth inhibition. These important pilot data will systematically introduce autophagy into translational AA research and is expected to identify a novel supportive therapeutic strategy in future AA management by nutraceuticals/cosmetics that targets autophagy.

Read more

Impact: If successful, this study may help inform new treatment strategies in alopecia areata through the use commercially available nutraceuticals that target autophagy.

A Novel Treg Augmenting Therapy For Patients With Alopecia Areata

Year: 2019 PI Name: Michael D. Rosenblum, MD Award Type: Research Grant Status: Completed Summary:
This project will examine the function of regulatory T cells (Tregs) in the activation and differentiation of hair follicle stem cells in alopecia areata and determine whether the activity of Tregs can be modulated in alopecia areata using a novel IL-2 molecule.

Read more

Abstract:
Regulatory T cells (Tregs) play a major role in establishing and maintaining immune homeostasis. We have discovered that both murine and human skin contain a unique population of Tregs that preferentially localize to hair follicles (HFs) and that these cells are required for HF regeneration as well as proper HF cycling. In addition, we have found that Tregs mediate these effects by promoting the activation and differentiation of hair follicle stem cells (HFSCs). The underlying theme of this proposal is to provide the cellular and molecular foundation for Treg augmentation therapy to treat patients with alopecia areata (AA). To achieve this goal, we will assess whether augmenting Tregs in mice results in enhanced HFSC function and HF cycling/regeneration. In addition, using cutting-edge CyTOF technology, we will comprehensively quantify the immune cell subsets infiltrating AA skin. In final experiments, we will attempt to augment Tregs in single cell suspensions derived from lesional AA skin and Treg activation quantified relative to other immune cell subsets. Given that AA is defined by both a dysfunction in HF regeneration and a defect in Tregs, our results may have direct implications for patients suffering from this disease.

Read more

Impact: If successful, this study may help pave the way for future clinical trials testing the safety and efficacy of regulatory T cell (Treg) augmenting therapies in patients with alopecia areata.

Role Of Autophagy In Alopecia Areata

Year: 2019 PI Name: Rupali Gund, PhD Award Type: Mentored Investigator Award Status: Completed Summary:
This project aims to understand the role of autophagy (a normal physiological process in the body that deals with destruction of cells in the body) in normal hair cycling and its role in the development of alopecia areata in the C3H/HeJ mouse model.

Read more

Abstract:
Alopecia Areata (AA) is an organ-restricted autoimmune disease that specifically attacks the hair follicles, resulting in well-demarcated (AA Patchy) or diffuse non-scarring hair loss of the scalp (AA Totalis) or the entire body (AA Universalis). AA is a highly prevalent disease with a lifetime risk of 2.1%, however, the underlying disease mechanisms remain incompletely defined and under-studied. Histologically, AA presents as a “swarm of bees” in which inflammatory T lymphocytes attack the pigmented actively growing hair follicles. The prevalent notion for disease is thought to be the loss of immune privilege of hair follicles causing abnormal activation of pathogenic T cells. Genetic association studies previously conducted in our lab found several autophagy related genes associated with AA. In addition, our gene expression analysis revealed altered expression of autophagy related genes, prompting us to hypothesize that dysregulation of autophagy plays a critical role in AA pathogenesis. Autophagy is a survivalpromoting mechanism that involves capturing, degradation, and recycling of intracellular proteins and organelles in lysosomes. However, the contribution of autophagy in loss of hair follicle immune privilege and development of AA is not characterized. This project aims to understand the role of autophagy in normal hair cycling and its contribution to the development of alopecia in grafted C3H/HeJ mouse model.

Read more

Impact: If successful, this study could provide new insights into the mechanism of alopecia areata pathogenesis mediated by autophagy and also identify novel targets for therapeutic intervention.