Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Publication year range
1.
N Engl J Med ; 386(18): 1687-1699, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35334197

ABSTRACT

BACKGROUND: Alopecia areata is an autoimmune condition characterized by rapid hair loss in the scalp, eyebrows, and eyelashes, for which treatments are limited. Baricitinib, an oral, selective, reversible inhibitor of Janus kinases 1 and 2, may interrupt cytokine signaling implicated in the pathogenesis of alopecia areata. METHODS: We conducted two randomized, placebo-controlled, phase 3 trials (BRAVE-AA1 and BRAVE-AA2) involving adults with severe alopecia areata with a Severity of Alopecia Tool (SALT) score of 50 or higher (range, 0 [no scalp hair loss] to 100 [complete scalp hair loss]). Patients were randomly assigned in a 3:2:2 ratio to receive once-daily baricitinib at a dose of 4 mg, baricitinib at a dose of 2 mg, or placebo. The primary outcome was a SALT score of 20 or less at week 36. RESULTS: We enrolled 654 patients in the BRAVE-AA1 trial and 546 in the BRAVE-AA2 trial. The estimated percentage of patients with a SALT score of 20 or less at week 36 was 38.8% with 4-mg baricitinib, 22.8% with 2-mg baricitinib, and 6.2% with placebo in BRAVE-AA1 and 35.9%, 19.4%, and 3.3%, respectively, in BRAVE-AA2. In BRAVE-AA1, the difference between 4-mg baricitinib and placebo was 32.6 percentage points (95% confidence interval [CI], 25.6 to 39.5), and the difference between 2-mg baricitinib and placebo was 16.6 percentage points (95% CI, 9.5 to 23.8) (P<0.001 for each dose vs. placebo). In BRAVE-AA2, the corresponding values were 32.6 percentage points (95% CI, 25.6 to 39.6) and 16.1 percentage points (95% CI, 9.1 to 23.2) (P<0.001 for each dose vs. placebo). Secondary outcomes for baricitinib at a dose of 4 mg but not at a dose of 2 mg generally favored baricitinib over placebo. Acne, elevated levels of creatine kinase, and increased levels of low- and high-density lipoprotein cholesterol were more common with baricitinib than with placebo. CONCLUSIONS: In two phase 3 trials involving patients with severe alopecia areata, oral baricitinib was superior to placebo with respect to hair regrowth at 36 weeks. Longer trials are required to assess the efficacy and safety of baricitinib for alopecia areata. (Funded by Eli Lilly under license from Incyte; BRAVE-AA1 and BRAVE-AA2 ClinicalTrials.gov numbers, NCT03570749 and NCT03899259.).


Subject(s)
Alopecia Areata , Janus Kinase Inhibitors , Adult , Alopecia Areata/drug therapy , Azetidines/adverse effects , Azetidines/therapeutic use , Humans , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/therapeutic use , Purines/adverse effects , Purines/therapeutic use , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Sulfonamides/adverse effects , Sulfonamides/therapeutic use
2.
Nat Immunol ; 13(8): 744-52, 2012 Jun 24.
Article in English | MEDLINE | ID: mdl-22729248

ABSTRACT

Langerhans cells (LCs) are epidermal dendritic cells with incompletely understood origins that associate with hair follicles for unknown reasons. Here we show that in response to external stress, mouse hair follicles recruited Gr-1(hi) monocyte-derived precursors of LCs whose epidermal entry was dependent on the chemokine receptors CCR2 and CCR6, whereas the chemokine receptor CCR8 inhibited the recruitment of LCs. Distinct hair-follicle regions had differences in their expression of ligands for CCR2 and CCR6. The isthmus expressed the chemokine CCL2; the infundibulum expressed the chemokine CCL20; and keratinocytes in the bulge produced the chemokine CCL8, which is the ligand for CCR8. Thus, distinct hair-follicle keratinocyte subpopulations promoted or inhibited repopulation with LCs via differences in chemokine production, a feature also noted in humans. Pre-LCs failed to enter hairless skin in mice or humans, which establishes hair follicles as portals for LCs.


Subject(s)
Chemokines/biosynthesis , Hair Follicle/immunology , Langerhans Cells/physiology , Stress, Physiological , Alopecia , Animals , Cell Movement , Chemokine CCL20/biosynthesis , Chemokine CCL8/biosynthesis , Chemokines/metabolism , Hair Follicle/metabolism , Humans , Keratinocytes/metabolism , Langerhans Cells/immunology , Mice , Mice, Hairless , Receptors, CCR2/metabolism , Receptors, CCR6/metabolism , Receptors, CCR8/metabolism , Skin/immunology
3.
Br J Dermatol ; 189(6): 666-673, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37708254

ABSTRACT

BACKGROUND: Baricitinib is approved for the treatment of adults with severe alopecia areata (AA). In the absence of robust data on the patterns of regrowth during treatment of severe AA, there is a gap in the knowledge regarding treatment expectations. OBJECTIVES: To examine whether different clinical response subgroups could be identified in baricitinib-treated patients with severe AA and factors that contribute to these subgroups. METHODS: The BRAVE-AA1 and BRAVE-AA2 phase III trials enrolled patients with severe AA [Severity of Alopecia Tool (SALT) score ≥ 50 (≥ 50% scalp hair loss)]. Patients randomized to baricitinib 4 mg or 2 mg retained their treatment allocation for 52 weeks. Based on patterns identified through growth mixture modelling (GMM), patients were categorized into responder subgroups according to when they first achieved ≥ 30% improvement from baseline in SALT score (SALT30). For each responder subgroup, trajectories of response (i.e. achievement of a SALT score ≤ 20, SALT score ≤ 10 and ≥ 50% change from baseline in SALT score) and baseline disease characteristics are reported. RESULTS: Respectively, 515 and 340 patients were randomized to once-daily baricitinib 4 mg and 2 mg at baseline; 69% and 51%, respectively, achieved SALT30 at least once by week 52. Based on GMM findings, we identified three responder subgroups: early (SALT30 by week 12), gradual (SALT30 after week 12-week 36) and late (SALT30 after week 36-week 52). The proportions of early, gradual and late responders and nonresponders were, respectively, 33%, 28%, 8% and 31% among patients treated with baricitinib 4 mg, and 20%, 23%, 9% and 49%, respectively, among those treated with baricitinib 2 mg. Early responders had a shorter trajectory to maximal clinical outcomes (e.g. > 78% achieved a SALT score ≤ 20 by week 36) vs. gradual or late responders. Early responders were more frequent among patients with baseline severe AA (SALT score 50 to < 95) vs. very severe AA (SALT score 95-100). Overall, responders (early to late) were more frequent in patients with short (< 4 years) episodes of hair loss. CONCLUSIONS: These analyses identified early, gradual and late responder subgroups for scalp hair regrowth in baricitinib-treated patients with severe AA, and that these subgroups are influenced by baseline characteristics. Findings from these analyses will help to inform treatment expectations for scalp hair regrowth.


Subject(s)
Alopecia Areata , Azetidines , Purines , Pyrazoles , Sulfonamides , Adult , Humans , Alopecia Areata/drug therapy , Hair , Scalp , Randomized Controlled Trials as Topic , Clinical Trials, Phase III as Topic
4.
Int J Mol Sci ; 23(13)2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35806129

ABSTRACT

In a clinical study of autologous cell-based therapy using dermal sheath cup (DSC) cells, the treatment of hair loss showed improvements. However, the outcomes were variable. Here, correlations between marker gene expression in DSC cells and treatment outcomes were assessed to predict therapeutic efficacy. Overall, 32 DSC cell lines were used to evaluate correlations between marker gene expression and treatment outcomes. Correlations between vascular pericyte and preadipocyte marker expression and treatment outcomes were inconsistent. As smooth muscle cell markers, MYOCD correlated negatively with treatment outcomes and SRF consistently demonstrated an inverse correlation. Additionally, CALD1 correlated negatively and ACTA2 correlated inversely with treatment outcomes. DSC cell lines were divided into good and moderate/poor responders to further investigate the correlations. SRF and CALD1 were lower in a good responder compared with a moderate responder. Next, DSC cells were differentiated toward dermal papilla cells. Dermal papilla markers SOX2 and LEF1 before differentiation had moderate positive and inverse correlations with the treatment outcome, respectively. SOX2 after differentiation more consistently demonstrated a positive correlation. Significant downregulation of smooth muscle-related genes was also observed after differentiation. These findings revealed putative markers for preclinical evaluation of DSC cells to improve hair loss.


Subject(s)
Alopecia , Hair Follicle , Alopecia/genetics , Alopecia/metabolism , Alopecia/therapy , Biomarkers/metabolism , Cells, Cultured , Female , Hair Follicle/metabolism , Humans , Male , Myocytes, Smooth Muscle/metabolism , Skin/metabolism , Treatment Outcome
5.
J Am Acad Dermatol ; 85(4): 847-853, 2021 10.
Article in English | MEDLINE | ID: mdl-34090959

ABSTRACT

BACKGROUND: There are no treatments approved by the Food and Drug Administration for alopecia areata. OBJECTIVE: To evaluate the efficacy and safety of baricitinib in patients with ≥50% scalp hair loss in a phase 2 study of adults with alopecia areata (BRAVE-AA1). METHODS: Patients were randomized 1:1:1:1 to receive placebo or baricitinib 1 mg, 2 mg, or 4 mg once daily. Two consecutive interim analyses were performed after all patients completed weeks 12 and 36 or had discontinued treatment prior to these time points. The primary endpoint was the proportion of patients achieving a Severity of Alopecia Tool (SALT) score ≤20 at week 36. Logistic regression was used with nonresponder imputation for missing data. RESULTS: A total of 110 patients were randomized (placebo, 28; baricitinib 1-mg, 28; 2-mg, 27; 4-mg, 27). The baricitinib 1-mg dose was dropped after the first interim analysis based on lower SALT30 response rate. At week 36, the proportion of patients achieving a SALT score of ≤20 was significantly greater in baricitinib 2-mg (33.3%, P = .016) and 4-mg (51.9%, P = .001) groups versus placebo (3.6%). Baricitinib was well tolerated with no new safety findings. LIMITATIONS: Small sample size limits generalizability of results. CONCLUSION: These results support the efficacy and safety of baricitinib in patients with ≥50% scalp hair loss.


Subject(s)
Alopecia Areata , Janus Kinase Inhibitors , Adult , Alopecia Areata/drug therapy , Azetidines , Humans , Janus Kinase Inhibitors/adverse effects , Purines , Pyrazoles , Sulfonamides , Treatment Outcome
6.
Int J Clin Oncol ; 25(10): 1861-1869, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32656742

ABSTRACT

BACKGROUND: The blockade of cell surface PD-1 ((sur)PD-1) by monoclonal antibodies, represented by nivolumab, provides the strategy to treat advanced malignant melanoma (AMM). The intracellular presence of PD-1 molecules have been reported in some T cell subsets, however, their kinetic association with those expressed on the cell surface, let alone their significance in antitumor immunity has been ill-investigated. METHODS: Intracellular PD-1 expression status in T cell subsets in AMM cases during nivolumab administration was chronologically characterized. The kinetics of PD-1 molecules within AMM-derived T cells was assessed in vitro in conjunction with their functional properties. RESULTS: Increase in (sur)PD-1 and intracellular PD-1 ((int)PD-1+) expression was characteristic for AMM T cells. After short-term culture, virtually (sur)PD-1- nivolumab-treated AMM T cells restore (sur)PD-1 expression, which could not be explained by the detachment of nivolumab from PD-1 epitopes alone. The blockade of trans-Golgi network resulted in the decrease in the extent of (sur)PD-1 recovery, suggesting the translocation of accumulated (int)PD-1 to the cell surface. Antigen-specific PD-1+ T cells significantly increased in (int)PD-1+ cells after treatment. In addition, a surge in (int)PD-1+CD4+ T cells was observed prior to the emergence of skin rash as an immune-related adverse event (irAE). CONCLUSIONS: Accumulated (int)PD-1 in T cells may contribute to enhanced immune evasion in AMM. Evaluation of intracellular PD-1 expression would be useful for better management of nivolumab-treated AMM patients in view of predicting treatment response and the incidence of irAE. Our findings further support the necessity of periodical administration of nivolumab for treating AMM.


Subject(s)
Melanoma/immunology , Nivolumab/pharmacology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/metabolism , Tumor Escape/immunology , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Humans , Immunotherapy/adverse effects , Melanoma/drug therapy , Melanoma/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Escape/drug effects
8.
Vet Dermatol ; 30(5): 365-e107, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31297916

ABSTRACT

BACKGROUND: Nestin, which was originally described as a neural crest stem cell marker, is known to be expressed in bulge follicle cells of human, canine and murine anagen hairs. However, the capacity of nestin-expressing cells to differentiate into the components of the hair follicle or the epidermis has been insufficiently investigated. HYPOTHESIS/OBJECTIVES: To determine whether nestin-expressing cells are capable of differentiating into keratinocytes. ANIMALS/MATERIALS: A double-transgenic mouse line Nes-Cre/CAG-CAT-EGFP, in which enhanced green fluorescent protein (EGFP) is expressed upon Cre-based recombination driven by the nestin promoter. METHODS AND MATERIALS: The tissue distribution of EGFP+ and nestin+ cells in the skin of the mouse line was analysed by immunofluorescence and immunohistochemical analyses. RESULTS: EGFP+ cells were recognized in the outer epithelial cell layers of anagen and telogen hair follicles, but rarely seen in the interfollicular epidermis. The EGFP+ cells in the outer layers of the hair follicles coexpressed keratin 14, a marker of the outer root sheath (ORS) keratinocytes, but not trichohyalin granules, an inner root sheath keratinocyte cell marker. Immunostaining for nestin failed to detect its expression in the majority of hair follicle epithelial cells, suggesting that the EGFP+ cells in the ORS were derived from nestin-expressing progenitor cells that had become further committed along the epithelial cell lineage, where nestin is no longer expressed. CONCLUSIONS AND CLINICAL IMPORTANCE: These results suggest that progenitor cells that differentiate into ORS keratinocytes are distinct from those for other hair follicle or epidermal components and provide implications for regenerative medicine and the molecular classification of hair follicle tumours.


Subject(s)
Cell Differentiation/physiology , Hair Follicle/cytology , Keratinocytes/classification , Nestin/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Keratinocytes/physiology , Mice , Mice, Transgenic , Nestin/genetics
9.
Hum Mol Genet ; 24(17): 4879-900, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26056228

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is the causative molecule of the autosomal dominant hereditary form of Parkinson's disease (PD), PARK8, which was originally defined in a study of a Japanese family (the Sagamihara family) harboring the I2020T mutation in the kinase domain. Although a number of reported studies have focused on cell death mediated by mutant LRRK2, details of the pathogenetic effect of LRRK2 still remain to be elucidated. In the present study, to elucidate the mechanism of neurodegeneration in PD caused by LRRK2, we generated induced pluripotent stem cells (iPSC) derived from fibroblasts of PD patients with I2020T LRRK2 in the Sagamihara family. We found that I2020T mutant LRRK2 iPSC-derived neurons released less dopamine than control-iPSC-derived neurons. Furthermore, we demonstrated that patient iPSC-derived neurons had a lower phospho-AKT level than control-iPSC-derived neurons, and that the former showed an increased incidence of apoptosis relative to the controls. Interestingly, patient iPSC-derived neurons exhibited activation of glycogen synthase kinase-3ß (GSK-3ß) and high Tau phosphorylation. In addition, the postmortem brain of the patient from whom the iPSC had been established exhibited deposition of neurofibrillary tangles as well as increased Tau phosphorylation in neurons. These results suggest that I2020T LRRK2-iPSC could be a promising new tool for reproducing the pathology of PD in the brain caused by the I2020T mutation, and applicable as a model in studies of targeted therapeutics.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , tau Proteins/metabolism , Animals , Apoptosis/genetics , Autophagy , Caspase 3/metabolism , Cell Line , Dopamine/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Induced Pluripotent Stem Cells/cytology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Neurons/cytology , Oxidative Stress , Phosphorylation
10.
J Cutan Pathol ; 44(10): 861-866, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28632903

ABSTRACT

Panfolliculoma (PF) is a rare benign tumor with signs of differentiation toward all components of the hair follicle (HF). Cystic panfolliculoma (CPF) is a subset of PF with histological similarity to trichofolliculoma making the differential diagnosis difficult in some cases. Immunohistopathological investigations of PF have been reported; however, previous studies focused mostly on the expression profile of the outer root sheath markers. Herein, we report a case of CPF. A panel of antibodies was developed and used for the detection of the expression of cytokeratin (CK) 10, CK13, CK14, CK15, hair-hard keratin (AE13) and EpCAM within the lesion, supporting the differentiation of all epithelial lineages of the HF and the diagnosis of CPF. Immunohistopathological examinations clearly showed the spatial distribution pattern of each subset of tumor cells with distinct HF differentiation marker expression. Intriguingly, fibroblastic dermal cells were distributed preferentially near CK15-negative epithelium or CK13-positive HF-like structures, suggesting a role for epithelial-mesenchymal interactions (EMIs) in CPF pathogenesis. Further characterization of EMIs between the tumor and surrounding mesenchymal cells in CPF may provide an explanation for immature HF differentiation. These findings suggest that the more intense and coordinated EMI in the analogous tumorigenesis gives rise to mature HF structures, resulting in trichofolliculoma, which may explain their histological resemblance.


Subject(s)
Gene Expression Regulation, Neoplastic , Hair Follicle , Keratins/biosynthesis , Neoplasm Proteins/biosynthesis , Skin Neoplasms , Aged , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Male , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
11.
Differentiation ; 92(5): 270-280, 2016 12.
Article in English | MEDLINE | ID: mdl-27178356

ABSTRACT

Mesenchymal stem cells (MSCs) are defined as non-hematopoietic, plastic-adherent, self-renewing cells that are capable of tri-lineage differentiation into bone, cartilage or fat in vitro. Thus, MSCs are promising candidates for cell-based medicine. However, classifications of MSCs have been defined retrospectively; moreover, this conventional criterion may be inaccurate due to contamination with other hematopoietic lineage cells. Human MSCs can be enriched by selection for LNGFR and THY-1, and this population may be analogous to murine PDGFRα+Sca-1+ cells, which are developmentally derived from neural crest cells (NCCs). Murine NCCs were labeled by fluorescence, which provided definitive proof of neural crest lineage, however, technical considerations prevent the use of a similar approach to determine the origin of human LNGFR+THY-1+ MSCs. To further clarify the origin of human MSCs, human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) were used in this study. Under culture conditions required for the induction of neural crest cells, human ESCs and iPSCs-derived cells highly expressed LNGFR and THY-1. These LNGFR+THY-1+ neural crest-like cells, designated as LT-NCLCs, showed a strong potential to differentiate into both mesenchymal and neural crest lineages. LT-NCLCs proliferated to form colonies and actively migrated in response to serum concentration. Furthermore, we transplanted LT-NCLCs into chick embryos, and traced their potential for survival, migration and differentiation in the host environment. These results suggest that LNGFR+THY-1+ cells identified following NCLC induction from ESCs/iPSCs shared similar potentials with multipotent MSCs.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Nerve Tissue Proteins/genetics , Receptors, Nerve Growth Factor/genetics , Thy-1 Antigens/genetics , Animals , Cell Culture Techniques , Cell Lineage/genetics , Cell Proliferation/genetics , Chick Embryo , Human Embryonic Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Mice , Neural Crest/cytology , Neural Crest/growth & development
14.
Proc Natl Acad Sci U S A ; 110(4): 1434-9, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23292936

ABSTRACT

Smoothened antagonists directly target the genetic basis of human basal cell carcinoma (BCC), the most common of all cancers. These drugs inhibit BCC growth, but they are not curative. Although BCC cells are monomorphic, immunofluorescence microscopy reveals a complex hierarchical pattern of growth with inward differentiation along hair follicle lineages. Most BCC cells express the transcription factor KLF4 and are committed to terminal differentiation. A small CD200(+) CD45(-) BCC subpopulation that represents 1.63 ± 1.11% of all BCC cells resides in small clusters at the tumor periphery. By using reproducible in vivo xenograft growth assays, we determined that tumor initiating cell frequencies approximate one per 1.5 million unsorted BCC cells. The CD200(+) CD45(-) BCC subpopulation recreated BCC tumor growth in vivo with typical histological architecture and expression of sonic hedgehog-regulated genes. Reproducible in vivo BCC growth was achieved with as few as 10,000 CD200(+) CD45(-) cells, representing ~1,500-fold enrichment. CD200(-) CD45(-) BCC cells were unable to form tumors. These findings establish a platform to study the effects of Smoothened antagonists on BCC tumor initiating cell and also suggest that currently available anti-CD200 therapy be considered, either as monotherapy or an adjunct to Smoothened antagonists, in the treatment of inoperable BCC.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Basal Cell/immunology , Carcinoma, Basal Cell/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Animals , Carcinoma, Basal Cell/metabolism , Cell Differentiation , Cell Proliferation , Humans , Keratins/metabolism , Kruppel-Like Factor 4 , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Receptors, G-Protein-Coupled/antagonists & inhibitors , Skin Neoplasms/metabolism , Smoothened Receptor , Transplantation, Heterologous , Tumor Stem Cell Assay
15.
J Immunol ; 191(1): 83-90, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23729442

ABSTRACT

Paraneoplastic pemphigus (PNP) is an autoimmune disease of the skin and mucous membranes that can involve fatal lung complications. IgG autoantibodies target the cell adhesion molecules desmoglein (Dsg)3 and plakins, but the nature and targets of infiltrating T cells are poorly characterized. Moreover, the lung involvement in this skin Ag-specific autoimmune condition represents a paradox. To mimic autoimmunity in PNP, we grafted wild-type skin onto Dsg3(-/-) mice, which resulted in graft rejection and generation of anti-Dsg3 IgG and Dsg3-specific T cells. Transfer of splenocytes from these mice into Rag2(-/-) mice induced a combination of suprabasilar acantholysis and interface dermatitis, a histology unique to PNP. Furthermore, the recipient mice showed prominent bronchial inflammation of CD4(+) and CD8(+) T cells with high mortality. Intriguingly, ectopic Dsg3 expression was observed in the lungs of PNP mice, mirroring the observation that squamous metaplasia is often found in the lungs of PNP patients. Dsg3 and other epidermal Ags were ectopically expressed in the lungs after pulmonary injuries by naphthalene, which was sufficient for recruitment of Dsg3-specific CD4(+) T cells. These findings demonstrate that squamous metaplasia after pulmonary epithelial injury may play a crucial role in redirecting the skin-specific autoimmune reaction to the lungs in PNP.


Subject(s)
Desmoglein 3/biosynthesis , Epidermis/immunology , Lung/immunology , Paraneoplastic Syndromes/immunology , Pemphigus/immunology , Respiratory Mucosa/immunology , Animals , Autoantigens/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Desmoglein 3/deficiency , Desmoglein 3/immunology , Disease Models, Animal , Epidermis/metabolism , Epidermis/pathology , Humans , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organs at Risk , Paraneoplastic Syndromes/metabolism , Paraneoplastic Syndromes/pathology , Pemphigus/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
16.
J Cell Sci ; 125(Pt 17): 4114-25, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22623722

ABSTRACT

The dermal papilla (DP) plays pivotal roles in hair follicle morphogenesis and cycling. However, characterization and/or propagation of human DPs have been unsatisfactory because of the lack of efficient isolation methods and the loss of innate characteristics in vitro. We hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion of functional DP cells. To test this, we first characterized the global gene expression profile of microdissected, non-cultured human DPs. We performed a 'two-step' microarray analysis to exclude the influence of unwanted contaminants in isolated DPs and successfully identified 118 human DP signature genes, including 38 genes listed in the mouse DP signature. The bioinformatics analysis of the DP gene list revealed that WNT, BMP and FGF signaling pathways were upregulated in intact DPs and addition of 6-bromoindirubin-3'-oxime, recombinant BMP2 and basic FGF to stimulate these respective signaling pathways resulted in maintained expression of in situ DP signature genes in primarily cultured human DP cells. More importantly, the exposure to these stimulants restored normally reduced DP biomarker expression in conventionally cultured DP cells. Cell growth was moderate in the newly developed culture medium. However, rapid DP cell expansion by conventional culture followed by the restoration by defined activators provided a sufficient number of DP cells that demonstrated characteristic DP activities in functional assays. The study reported here revealed previously unreported molecular mechanisms contributing to human DP properties and describes a useful technique for the investigation of human DP biology and hair follicle bioengineering.


Subject(s)
Dermis/metabolism , Hair Follicle/metabolism , Animals , Cells, Cultured , Computational Biology , Culture Media/pharmacology , Dermis/cytology , Dermis/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Hair Follicle/cytology , Hair Follicle/drug effects , Humans , Mice , Microdissection , Oligonucleotide Array Sequence Analysis , RNA/genetics , RNA/isolation & purification , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Australas J Dermatol ; 55(2): 145-8, 2014 May.
Article in English | MEDLINE | ID: mdl-23714002

ABSTRACT

Cronkhite-Canada syndrome (CCS) is a rare disorder characterised by gastrointestinal polyposis and ectodermal changes, represented by extensive alopecia. Detailed histopathological investigations of alopecic lesions in two female CCS patients with severe hair loss revealed a marked increase in telogen hair follicles with no sign of loss or of the minaturisation or atrophy of hair follicle structures and the absence of inflammatory change, despite severe inflammation in the gastrointestinal tract. These findings suggested that hair regrowth can be expected without systemic corticosteroids, if they are not necessary for treatment of the gastrointestinal tract, and that anagen-telogen transition is an early event preceding clinical hair loss in CCS.


Subject(s)
Alopecia/pathology , Hair Follicle/pathology , Intestinal Polyposis/pathology , Antineoplastic Agents, Hormonal/therapeutic use , Disease Progression , Female , Hair Follicle/growth & development , Humans , Hypoalbuminemia/blood , Intestinal Polyposis/blood , Intestinal Polyposis/drug therapy , Middle Aged , Prednisone/therapeutic use
19.
Australas J Dermatol ; 55(1): e12-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23094684

ABSTRACT

We report a 78-year-old woman with rheumatoid neutrophilic dermatosis (RND) presenting with tense blisters; an extremely rare manifestation of this condition. Systemic corticosteroid was of limited efficacy, while dapsone was effective. A literature review of four similar cases showed that tense blisters in this type of RND tended to appear on the lower extremities of aged, female rheumatoid arthritis patients. Of note, half of the cases were resistant to corticosteroids, as anti-neutrophil agents are reported to be effective. Accordingly, it is important to recognise this unusual manifestation for the timely initiation of appropriate therapy.


Subject(s)
Arthritis, Rheumatoid/complications , Blister/etiology , Dermatitis/complications , Dermatitis/pathology , Aged , Anti-Infective Agents/therapeutic use , Blister/drug therapy , Dapsone/therapeutic use , Female , Humans , Neutrophil Infiltration
20.
Dermatol Ther (Heidelb) ; 14(1): 151-167, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38079099

ABSTRACT

INTRODUCTION: Alopecia areata (AA) is characterized by non-scarring scalp and/or body hair loss and can negatively impact patient mental health. Data are limited on the alignment of patient and physician perceptions of AA severity with each other and with Japanese Dermatological Association (JDA) guideline criteria, and of patient-physician alignment on treatment satisfaction. Therefore, we performed analyses to compare JDA severity groupings with patient-physician alignment on disease severity and to explore treatment satisfaction in AA in Japan. METHODS: Data were drawn from the Adelphi AA Disease Specific Programme (DSP)™, a real-world survey of physicians and patients with AA in Japan conducted January-March 2021. Patients and physicians reported patient AA severity as mild, moderate or severe based on their subjective judgement. Patients were then categorized into five hair loss severity groups according to JDA criteria (S1-5), and patient-physician pairs were matched to assess alignment on severity and treatment satisfaction. RESULTS: Subjective patient- and physician-reported disease severity generally followed JDA severity groupings. The percentage of patient-physician alignment on severity recognition was 76.3% in the overall population. In misaligned pairs, 20.2%, 14.5%, 7.3%, 25.0% and 0.0% of physicians rated disease as more severe than patients in S1, S2, S3, S4 and S5, respectively. Regarding treatment satisfaction, patient-physician alignment was 57.6% in the overall population. In S5, 46.2% of physicians reported being less satisfied than patients. Both physicians and patients cited lack of efficacy as the main reason for dissatisfaction. Of 221 patients, 39.8% and 29.9% were categorized as borderline-abnormal cases for anxiety and depression, respectively. CONCLUSIONS: This study highlights previously unreported patient-physician misalignment on disease severity, level of treatment dissatisfaction and unmet needs due to the lack of effective treatment. Further study on how improvement of the misalignment between physicians and patients could increase both patient and physician satisfaction with treatment and improve the quality of life for patients with AA.

SELECTION OF CITATIONS
SEARCH DETAIL