Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Dermatol Sci ; 114(1): 13-23, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448341

ABSTRACT

BACKGROUND: The aberrant expression of tight junction (TJ) proteins play an important role in several diseases with impaired skin barriers, including atopic dermatitis, psoriasis, and chronic wounds. The evidence provided thus far suggests an important role of calcitriol in skin homeostasis. However, it is not known whether calcitriol improves the impaired skin barrier. OBJECTIVE: To investigate the effect of calcitriol on TJ barrier function in human primary keratinocytes. METHODS: Normal human primary keratinocytes were stimulated with calcitriol, and the expression of TJ-related proteins was measured by real-time PCR and Western blotting. Immunofluorescence was used to examine the intercellular distribution of TJ-related proteins. TJ barrier function was assessed by the transepithelial electrical resistance (TER) assay. RESULTS: We demonstrated that calcitriol increased the expression levels of TJ-related proteins, including claudin-4, claudin-7, occludin, and zonula occludens (ZO)- 1. Calcitriol enhanced the distribution of TJ-related proteins at cellcell borders and induced the phosphorylation of pathways involved in the regulation of TJ barrier function, such as atypical protein kinase C (aPKC), Ras-related C3 botulinum toxin substrate 1 (Rac1), phosphoinositide 3-kinase (PI3K), and protein kinase B (Akt), as evidenced by the effects of specific inhibitors on the above pathways. Indeed, we confirmed that calcitriol enhanced TER in keratinocyte monolayers. CONCLUSION: These findings showed that calcitriol could modify the expression of keratinocyte TJ proteins, contributing to the maintenance of homeostatic barrier function.


Subject(s)
Calcitriol , Epidermis , Keratinocytes , Tight Junctions , Humans , Calcitriol/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Cells, Cultured , Epidermis/drug effects , Epidermis/metabolism , Signal Transduction/drug effects , Phosphorylation/drug effects , Occludin/metabolism , Primary Cell Culture , Zonula Occludens-1 Protein/metabolism , Claudins/metabolism , Claudins/genetics , Electric Impedance
2.
Int J Mol Sci ; 24(11)2023 May 27.
Article in English | MEDLINE | ID: mdl-37298299

ABSTRACT

Atopic dermatitis and psoriasis are prevalent chronic inflammatory skin diseases that are characterized by dysfunctional skin barriers and substantially impact patients' quality of life. Vitamin D3 regulates immune responses and keratinocyte differentiation and improves psoriasis symptoms; however, its effects on atopic dermatitis remain unclear. Here, we investigated the effects of calcitriol, an active form of vitamin D3, on an NC/Nga mouse model of atopic dermatitis. We observed that the topical application of calcitriol decreased the dermatitis scores and epidermal thickness of NC/Nga mice with atopic dermatitis compared to untreated mice. In addition, both stratum corneum barrier function as assessed by the measurement of transepidermal water loss and tight junction barrier function as evaluated by biotin tracer permeability assay were improved following calcitriol treatment. Moreover, calcitriol treatment reversed the decrease in the expression of skin barrier-related proteins and decreased the expression of inflammatory cytokines such as interleukin (IL)-13 and IL-33 in mice with atopic dermatitis. These findings suggest that the topical application of calcitriol might improve the symptoms of atopic dermatitis by repairing the dysfunctional epidermal and tight junction barriers. Our results suggest that calcitriol might be a viable therapeutic agent for the treatment of atopic dermatitis in addition to psoriasis.


Subject(s)
Dermatitis, Atopic , Psoriasis , Mice , Animals , Dermatitis, Atopic/metabolism , Calcitriol/therapeutic use , Cholecalciferol/pharmacology , Quality of Life , Skin/metabolism , Cytokines/metabolism , Interleukin-13/metabolism , Psoriasis/drug therapy , Psoriasis/metabolism , Disease Models, Animal
3.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982275

ABSTRACT

The antimicrobial peptide derived from insulin-like growth factor-binding protein 5 (AMP-IBP5) exhibits antimicrobial activities and immunomodulatory functions in keratinocytes and fibroblasts. However, its role in regulating skin barrier function remains unclear. Here, we investigated the effects of AMP-IBP5 on the skin barrier and its role in the pathogenesis of atopic dermatitis (AD). 2,4-Dinitrochlorobenzene was used to induce AD-like skin inflammation. Transepithelial electrical resistance and permeability assays were used to investigate tight junction (TJ) barrier function in normal human epidermal keratinocytes and mice. AMP-IBP5 increased the expression of TJ-related proteins and their distribution along the intercellular borders. AMP-IBP5 also improved TJ barrier function through activation of the atypical protein kinase C and Rac1 pathways. In AD mice, AMP-IBP5 ameliorated dermatitis-like symptoms restored the expression of TJ-related proteins, suppressed the expression of inflammatory and pruritic cytokines, and improved skin barrier function. Interestingly, the ability of AMP-IBP5 to alleviate inflammation and improve skin barrier function in AD mice was abolished in mice treated with an antagonist of the low-density lipoprotein receptor-related protein-1 (LRP1) receptor. Collectively, these findings indicate that AMP-IBP5 may ameliorate AD-like inflammation and enhance skin barrier function through LRP1, suggesting a possible role for AMP-IBP5 in the treatment of AD.


Subject(s)
Dermatitis, Atopic , Humans , Animals , Mice , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Antimicrobial Peptides , Keratinocytes/metabolism , Inflammation/metabolism , Cytokines/metabolism , Disease Models, Animal , Lipoproteins, LDL/metabolism , Skin/metabolism
4.
J Invest Dermatol ; 143(5): 751-761.e7, 2023 05.
Article in English | MEDLINE | ID: mdl-36455652

ABSTRACT

Human cathelicidin LL-37 is a multifunctional antimicrobial peptide that exhibits antimicrobial and immunomodulatory activities. LL-37 regulates skin barrier function and was recently reported to activate autophagy in macrophages. Because autophagy deficiency is associated with skin diseases characterized by a dysfunctional epidermal barrier, we hypothesized that LL-37 might regulate the skin barrier through autophagy modulation. We showed that LL-37 activated autophagy in human keratinocytes and three-dimensional skin equivalent models as indicated by increases in LC3 puncta formation, decreases in p62, and autophagosome and autolysosome formation. LL-37‒induced autophagy was suppressed by P2X7 receptor, adenosine monophosphate‒activated protein kinase, and unc-51-like kinase 1 inhibitors, suggesting that the P2X7, adenosine monophosphate‒activated protein kinase, and unc-51-like kinase 1 pathways are involved. Moreover, LL-37 enhanced the phosphorylation of adenosine monophosphate‒activated protein kinase and unc-51-like kinase 1. In addition, LL-37‒mediated autophagy involves the mechanistic target of rapamycin and MAPK pathways. Interestingly, the LL-37‒induced distribution of tight junction proteins and improvement in the tight junction barrier were inhibited in autophagy-deficient keratinocytes and keratinocytes and skin models treated with autophagy inhibitors, indicating that the LL-37‒mediated tight junction barrier is associated with autophagy activation. Collectively, these findings suggest that LL-37 is a potential therapeutic target for skin diseases characterized by dysfunctional autophagy and skin barriers.


Subject(s)
Antimicrobial Cationic Peptides , Cathelicidins , Humans , Adenosine Monophosphate/metabolism , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/metabolism , Autophagy , Autophagy-Related Protein-1 Homolog/metabolism , Cathelicidins/pharmacology , Cathelicidins/metabolism , Keratinocytes/metabolism , Sirolimus , Signal Transduction
5.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36232814

ABSTRACT

Betacellulin (BTC) is a peptide ligand that belongs to the epidermal growth factor family, the members of which have been implicated in skin morphogenesis, homeostasis, repair, and angiogenesis; however, the role of BTC in the regulation of the skin barrier remains unknown. To examine the role of BTC in skin barrier function, we analyzed atopic dermatitis (AD) transcriptomic data from Gene Expression Omnibus (GEO) datasets, performed BTC immunohistochemistry using human skin tissues, and evaluated the effects of BTC on primary human keratinocytes by real-time PCR, Western blotting, and assay of the transepidermal electrical resistance (TER), a functional parameter to monitor the tight junction barrier. We found that the gene expression of BTC was downregulated in skin lesions from patients with AD, and this downregulated expression recovered following biological treatments. Consistently, the BTC protein levels were downregulated in the lesional skin of AD patients compared with the normal skin of healthy participants, suggesting that the BTC levels in skin might be a biomarker for the diagnosis and therapy of AD. Furthermore, in human keratinocytes, BTC knockdown reduced the levels of skin-derived antimicrobial peptides and skin barrier-related genes, whereas BTC addition enhanced their levels. Importantly, in human skin equivalents, BTC restored the increased tight junction permeability induced by Th2 cytokine IL-4/IL-13 treatment. In addition, specific inhibitors of epidermal growth factor receptor (EGFR) and protein kinase C (PKC) abolished the BTC-mediated improvement in skin barrier-related proteins in keratinocyte monolayers. Collectively, our findings suggest that treatment with BTC might improve the Th2-type cytokine-mediated impairment of skin barrier function through the EGFR/PKC axis and that BTC might be a novel potential biomarker and therapeutic target for the treatment of skin conditions characterized by the overproduction of Th2 cytokines and dysfunctional skin barriers, such as AD.


Subject(s)
Cytokines , Dermatitis, Atopic , Betacellulin/metabolism , Cytokines/metabolism , Dermatitis, Atopic/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Interleukin-13/metabolism , Interleukin-13/pharmacology , Interleukin-4/metabolism , Keratinocytes/metabolism , Ligands , Protein Kinase C/metabolism , Skin/metabolism
6.
Int J Mol Sci ; 23(15)2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35955934

ABSTRACT

The skin produces a plethora of antimicrobial peptides that not only show antimicrobial activities against pathogens but also exhibit various immunomodulatory functions. Human ß-defensins (hBDs) are the most well-characterized skin-derived antimicrobial peptides and contribute to diverse biological processes, including cytokine production and the migration, proliferation, and differentiation of host cells. Additionally, hBD-3 was recently reported to promote wound healing and angiogenesis, by inducing the expression of various angiogenic factors and the migration and proliferation of fibroblasts. Angiogenin is one of the most potent angiogenic factors; however, the effects of hBDs on angiogenin production in fibroblasts remain unclear. Here, we investigated the effects of hBDs on the secretion of angiogenin by human dermal fibroblasts. Both in vitro and ex vivo studies demonstrated that hBD-1, hBD-2, hBD-3, and hBD-4 dose-dependently increased angiogenin production by fibroblasts. hBD-mediated angiogenin secretion involved the epidermal growth factor receptor (EGFR), Src family kinase, c-Jun N-terminal kinase (JNK), p38, and nuclear factor-kappa B (NF-κB) pathways, as evidenced by the inhibitory effects of specific inhibitors for these pathways. Indeed, we confirmed that hBDs induced the activation of the EGFR, Src, JNK, p38, and NF-κB pathways. This study identified a novel role of hBDs in angiogenesis, through the production of angiogenin, in addition to their antimicrobial activities and other immunomodulatory properties.


Subject(s)
Anti-Infective Agents , beta-Defensins , Anti-Infective Agents/pharmacology , Antimicrobial Peptides , Cells, Cultured , ErbB Receptors , Fibroblasts/metabolism , Humans , NF-kappa B/metabolism , Ribonuclease, Pancreatic , beta-Defensins/metabolism
7.
J Clin Invest ; 132(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-35834333

ABSTRACT

Human ß-defensin-3 (hBD-3) exhibits antimicrobial and immunomodulatory activities; however, its contribution to autophagy regulation remains unclear, and the role of autophagy in the regulation of the epidermal barrier in atopic dermatitis (AD) is poorly understood. Here, keratinocyte autophagy was restrained in the skin lesions of patients with AD and murine models of AD. Interestingly, hBD-3 alleviated the IL-4- and IL-13-mediated impairment of the tight junction (TJ) barrier through keratinocyte autophagy activation, which involved aryl hydrocarbon receptor (AhR) signaling. While autophagy deficiency impaired the epidermal barrier and exacerbated inflammation, hBD-3 attenuated skin inflammation and enhanced the TJ barrier in AD. Importantly, hBD-3-mediated improvement of the TJ barrier was abolished in autophagy-deficient AD mice and in AhR-suppressed AD mice, suggesting a role for hBD-3-mediated autophagy in the regulation of the epidermal barrier and inflammation in AD. Thus, autophagy contributes to the pathogenesis of AD, and hBD-3 could be used for therapeutic purposes.


Subject(s)
Dermatitis, Atopic , beta-Defensins , Animals , Autophagy , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Keratinocytes/pathology , Mice , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , beta-Defensins/genetics , beta-Defensins/metabolism , beta-Defensins/therapeutic use
8.
Wound Repair Regen ; 30(2): 232-244, 2022 03.
Article in English | MEDLINE | ID: mdl-35092133

ABSTRACT

Impaired keratinocyte functions are major factors that are responsible for delayed diabetic wound healing. In addition to its antimicrobial activity, the antimicrobial peptide derived from insulin-like growth factor-binding protein 5 (AMP-IBP5) activates mast cells and promotes keratinocyte and fibroblast proliferation and migration. However, its effects on diabetic wound healing remain unclear. Human keratinocytes were cultured in normal or high glucose milieus. The production of angiogenic growth factor and cell proliferation and migration were evaluated. Wounds in normal and streptozotocin-induced diabetic mice were monitored and histologically examined. We found that AMP-IBP5 rescued the high glucose-induced attenuation of proliferation and migration as well as the production of angiogenin and vascular endothelial growth factors in keratinocytes. The AMP-IBP5-induced activity was mediated by the epidermal growth factor receptor, signal transducer and activator of transcription 1 and 3, and mitogen-activated protein kinase pathways, as indicated by the inhibitory effects of pathway-specific inhibitors. In vivo, AMP-IBP5 markedly accelerated wound healing, increased the expression of angiogenic factors and promoted vessel formation in both normal and diabetic mice. Overall, the finding that AMP-IBP5 accelerated diabetic wound healing by protecting against glucotoxicity and promoting angiogenesis suggests that AMP-IBP5 might be a potential therapeutic target for treating chronic diabetic wounds.


Subject(s)
Diabetes Mellitus, Experimental , Somatomedins , Animals , Mice , Antimicrobial Peptides , Cell Movement , Diabetes Mellitus, Experimental/metabolism , Glucose/pharmacology , Keratinocytes , Somatomedins/metabolism , Somatomedins/pharmacology , Wound Healing
9.
Front Immunol ; 12: 712781, 2021.
Article in English | MEDLINE | ID: mdl-34594328

ABSTRACT

In addition to its antimicrobial activity, the skin-derived antimicrobial peptide human ß-defensin-3 (hBD-3) promotes keratinocyte proliferation and migration to initiate the wound healing process; however, its effects on fibroblasts, which are the major cell type responsible for wound healing, remain unclear. We investigated the role of hBD-3 in cell migration, proliferation and production of angiogenic growth factors in human fibroblasts and evaluated the in vivo effect of hBD-3 on promoting wound healing and angiogenesis. Following hBD-3 treatment, the mouse wounds healed faster and showed accumulation of neutrophils and macrophages in the early phase of wound healing and reduction of these phagocytes 4 days later. hBD-3-treated wounds also displayed an increased number of fibroblasts and newly formed vessels compared to those of the control mice. Furthermore, the expression of various angiogenic growth factors was increased in the hBD-3-treated wounds. Additionally, in vitro studies demonstrated that hBD-3 enhanced the secretion of angiogenic growth factors such as fibroblast growth factor, platelet-derived growth factor and vascular endothelial growth factor and induced the migration and proliferation of human fibroblasts. The hBD-3-mediated activation of fibroblasts involves the fibroblast growth factor receptor 1 (FGFR1)/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathways, as evidenced by the inhibitory effects of pathway-specific inhibitors. We indeed confirmed that hBD-3 enhanced the phosphorylation of FGFR1, JAK2 and STAT3. Collectively, the current study provides novel evidence that hBD-3 might be a potential candidate for the treatment of wounds through its ability to promote wound healing, angiogenesis and fibroblast activation.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Antimicrobial Peptides/pharmacology , Cell Movement/drug effects , Signal Transduction/drug effects , Wound Healing/drug effects , beta-Defensins/pharmacology , Animals , Biomarkers , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Janus Kinase 2/metabolism , Matrix Metalloproteinase 2/metabolism , Mice , Models, Biological , Phosphorylation , Receptors, Fibroblast Growth Factor/metabolism , STAT3 Transcription Factor
10.
PLoS One ; 16(4): e0250663, 2021.
Article in English | MEDLINE | ID: mdl-33905439

ABSTRACT

In a disease-state-dependent manner, the histamine-resistant itch in dry skin-based skin diseases such as atopic dermatitis (AD) and xerosis is mainly due to hyperinnervation in the epidermis. Semaphorin 3A (Sema3A) is a nerve repulsion factor expressed in keratinocytes and it suppresses nerve fiber elongation in the epidermis. Our previous studies have shown that Sema3A ointment inhibits epidermal hyperinnervation and scratching behavior and improves dermatitis scores in AD model mice. Therefore, we consider Sema3A as a key therapeutic target for improving histamine-resistant itch in AD and xerosis. This study was designed to screen a library of herbal plant extracts to discover compounds with potential to induce Sema3A in normal human epidermal keratinocytes (NHEKs) using a reporter gene assay, so that positive samples were found. Among the positive samples, only the extract of S. baicalensis was found to consistently increase Sema3A levels in cultured NHEKs in assays using quantitative real-time PCR and ELISA. In evaluation of reconstituted human epidermis models, the level of Sema3A protein in culture supernatants significantly increased by application of the extract of S. baicalensis. In addition, we investigated which components in the extract of S. baicalensis contributed to Sema3A induction and found that baicalin and baicalein markedly increased the relative luciferase activity, and that baicalein had higher induction activity than baicalin. Thus, these findings suggest that S. baicalensis extract and its compounds, baicalin and baicalein, may be promising candidates for improving histamine-resistant itch via the induction of Sema3A expression in epidermal keratinocytes.


Subject(s)
Plant Extracts/chemistry , Scutellaria baicalensis/chemistry , Semaphorin-3A/metabolism , Cell Line , Flavanones/genetics , Flavanones/metabolism , Flavonoids/genetics , Flavonoids/metabolism , Genes, Reporter , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Models, Biological , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Scutellaria baicalensis/metabolism , Semaphorin-3A/genetics
11.
Biomedicines ; 9(3)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33668714

ABSTRACT

Itch or pruritus is the hallmark of atopic dermatitis and is defined as an unpleasant sensation that evokes the desire to scratch. It is also believed that itch is a signal of danger from various environmental factors or physiological abnormalities. Because histamine is a well-known substance inducing itch, H1-antihistamines are the most frequently used drugs to treat pruritus. However, H1-antihistamines are not fully effective against intractable itch in patients with atopic dermatitis. Given that intractable itch is a clinical problem that markedly decreases quality of life, its treatment in atopic dermatitis is of high importance. Histamine-independent itch may be elicited by various pruritogens, including proteases, cytokines, neuropeptides, lipids, and opioids, and their cognate receptors, such as protease-activated receptors, cytokine receptors, Mas-related G protein-coupled receptors, opioid receptors, and transient receptor potential channels. In addition, cutaneous hyperinnervation is partly involved in itch sensitization in the periphery. It is believed that dry skin is a key feature of intractable itch in atopic dermatitis. Treatment of the underlying conditions that cause itch is necessary to improve the quality of life of patients with atopic dermatitis. This review describes current insights into the pathophysiology of itch and its treatment in atopic dermatitis.

12.
Clin Exp Allergy ; 51(3): 382-392, 2021 03.
Article in English | MEDLINE | ID: mdl-33394511

ABSTRACT

Atopic dermatitis (AD) is a chronic relapsing inflammatory cutaneous disease that is often associated with other atopic symptoms, such as food allergy, allergic rhinitis and asthma, leading to significant morbidity and healthcare costs. The pathogenesis of AD is complicated and multifactorial. Although the aetiology of AD remains incompletely understood, recent studies have provided further insight into AD pathophysiology, demonstrating that the interaction among genetic predisposition, immune dysfunction and environmental provocation factors contributes to its development. However, the increasing prevalence of AD suggests that environmental factors such as irritation and cutaneous infection play a crucial role in triggering and/or aggravating the disease. Of note, AD skin is susceptible to bacterial, fungal and viral infections, and microorganisms may colonize the skin and aggravate AD symptoms. Overall, understanding the mechanisms by which these risk factors affect the cutaneous immunity of patients with AD is of great importance for developing a precision medicine approach for treatment. This review summarizes recent developments in exogenous factors involved in the pathogenesis of AD, with special emphasis on irritants and microbial infections.


Subject(s)
Dermatitis, Atopic/physiopathology , Irritants/adverse effects , Skin Diseases, Infectious/microbiology , Skin/microbiology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Humans , Kaposi Varicelliform Eruption/immunology , Kaposi Varicelliform Eruption/physiopathology , Microbiota , Molluscum Contagiosum/immunology , Molluscum Contagiosum/physiopathology , Skin Diseases, Infectious/immunology , Skin Diseases, Infectious/physiopathology
13.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066696

ABSTRACT

Atopic dermatitis (AD) is a common chronic inflammatory skin disease that exhibits a complex interplay of skin barrier disruption and immune dysregulation. Patients with AD are susceptible to cutaneous infections that may progress to complications, including staphylococcal septicemia. Although most studies have focused on filaggrin mutations, the physical barrier and antimicrobial barrier also play critical roles in the pathogenesis of AD. Within the physical barrier, the stratum corneum and tight junctions play the most important roles. The tight junction barrier is involved in the pathogenesis of AD, as structural and functional defects in tight junctions not only disrupt the physical barrier but also contribute to immunological impairments. Furthermore, antimicrobial peptides, such as LL-37, human b-defensins, and S100A7, improve tight junction barrier function. Recent studies elucidating the pathogenesis of AD have led to the development of barrier repair therapy for skin barrier defects in patients with this disease. This review analyzes the association between skin barrier disruption in patients with AD and antimicrobial peptides to determine the effect of these peptides on skin barrier repair and to consider employing antimicrobial peptides in barrier repair strategies as an additional approach for AD management.


Subject(s)
Cathelicidins/metabolism , Defensins/metabolism , Dermatitis, Atopic/metabolism , Skin/metabolism , Wound Healing , Dermatitis, Atopic/pathology , Filaggrin Proteins , Humans , Skin/pathology , Skin Physiological Phenomena
14.
Sci Rep ; 10(1): 4360, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152328

ABSTRACT

Because intractable itch reduces quality of life, understanding the fundamental mechanisms of itch is required to develop antipruritic treatments. Itch is mediated by peripheral sensory neurons, which originate from the neural crest (NC) during development. Itch-associated signaling molecules have been detected in genetically engineered animals and in cultures of peripheral neurons from dorsal root ganglia (DRG). Ethical difficulties collecting peripheral neurons from human DRG have limited analysis of itch in humans. This study describes a method of differentiating peripheral neurons from human induced pluripotent stem cells (hiPSCs) for physiological study of itch. This method resulted in the robust induction of p75 and HNK1 double-positive NC cells from hiPSCs. The expression of NC markers TFAP2A, SOX10 and SNAI1 increased during NC induction. The induction efficiency was nearly 90%, and human peripheral neurons expressing peripherin were efficiently differentiated from hiPSC-derived NC cells. Moreover, induced peripheral neurons expressed the sensory neuronal marker BRN3A and the itch-related receptors HRH1, MRGPRX1, IL31R and IL-4R. Calcium imaging analyses indicated that these peripheral neurons included sensory neurons responsive to itch-related stimuli such as histamine, BAM8-22, IL-31 and IL-4. These findings may enable detailed analyses of human DRG neurons and may result in new therapies for intractable itch.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Crest/cytology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Apoptosis , Biomarkers , Cell Differentiation/genetics , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/drug effects , Neurogenesis
15.
J Dermatol ; 47(4): 413-417, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31985094

ABSTRACT

Kakato-tsurutsuru (Kt) socks have been selling for almost 30 years in Japan. Wearers claim they improve heel dryness despite no scientific evidence. We investigated the effects of Kt socks on heel dryness by questionnaire, clinical scores and non-invasive skin measurements. In a double-blind, randomized cross-over study, 10 healthy volunteers wore control or Kt socks over 2 weeks in sequence for 4 weeks. Skin hydration and evaporation of the medial and dorsal heel were measured before and every week during the trial. Clinical evaluations of desquamation and cracked skin were scored by a dermatologist. A visual analog scale (VAS) questionnaire of comfort, sock climate humidity and skin dryness was conducted. The VAS of comfort was significantly higher in Kt than controls. Average Δskin dryness in control and Kt groups was -1.63 and 2.22, respectively, showing a significant improvement. In the clinical findings of the dorsal side of the heel, Δdesquamation and Δcracked skin scores were significantly decreased and Δstratum corneum hydration significantly increased in Kt compared with controls. Kt socks may retain evaporated sweat with components of natural moisturizing factors, supporting the water-holding ability of the heel stratum corneum. These findings suggest that Kt socks may improve heel skin dryness.


Subject(s)
Clothing , Emollients/administration & dosage , Epidermis/drug effects , Water Loss, Insensible/drug effects , Aged , Cross-Over Studies , Double-Blind Method , Epidermis/metabolism , Female , Healthy Volunteers , Heel , Humans , Japan , Male , Middle Aged , Treatment Outcome
16.
J Invest Dermatol ; 140(7): 1346-1354.e5, 2020 07.
Article in English | MEDLINE | ID: mdl-31945349

ABSTRACT

Epidermal keratinocytes are primarily involved in the expression of semaphorin (Sema) 3A, which is involved in the regulation of cutaneous innervation. However, the mechanisms underlying the intracellular signaling of Sema3A expression in keratinocytes remain unknown. We herein investigated the signaling mechanisms for the induction of Sema3A expression in normal human epidermal keratinocytes (NHEKs). Sema3A expression is transiently increased in calcium-stimulated NHEKs, whereas it is markedly decreased in terminally differentiated NHEKs. Sema3A mRNA is mainly localized in the stratum basale and stratum suprabasale of the epidermis. We cloned the 5'-flanking region of the Sema3A gene and identified a critical region for Sema3A promoter activity within -134 base pairs of the start codon. We found transcription factor binding sites, including that for activator protein (AP)-1, in this region. Sema3A expression was increased by the co-overexpression of JunB and Fra-2 in the presence of 0.1 or 1.4 mM calcium. The calcium-mediated transient upregulation of Sema3A expression was significantly suppressed by mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK) 1/2 or AP-1 inhibitors. These results demonstrate that the calcium-mediated transient upregulation of Sema3A in NHEKs is involved in the MEK/ERK and AP-1 signaling axis. Therefore, Sema3A mRNA may be expressed in the lower epidermis under controlled conditions by calcium via the MAPK-AP-1 axis.


Subject(s)
Keratinocytes/metabolism , MAP Kinase Signaling System , Semaphorin-3A/metabolism , Transcription Factor AP-1/metabolism , Binding Sites , Calcium/metabolism , Cell Differentiation , Cell Line , Epidermal Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Keratin-10/metabolism , Keratin-14/metabolism , RNA, Messenger/metabolism , Signal Transduction
20.
Acta Derm Venereol ; 97(8): 928-933, 2017 Aug 31.
Article in English | MEDLINE | ID: mdl-28512665

ABSTRACT

The pathogenesis of psoriatic itch is poorly understood. The aim of this study was to investigate the involvement of opioid receptors in scratching behaviour of imiquimod-induced psoriasis-like dermatitis model mice. Topical application of 5% imiquimod cream to the rostral back skin of mice induced antihistamine-resistant scratching behaviour. The expression of µ-opioid receptor (MOR) protein increased in the epidermis, dorsal root ganglia (DRG) and spinal cord of imiquimod-treated mice. In contrast, the expression of κ-opioid receptor (KOR) protein decreased in the DRG and spinal cord of imiquimod-treated mice, and was undetectable in the epidermis of both groups. Topical or intraperitoneal administration of the MOR antagonist naloxone and oral administration of the centrally acting KOR agonist ICI-199,441 inhibited scratching behaviour, whereas oral administration of the peri-pherally-selective KOR agonist asimadoline did not. These results suggest that peripheral and central MOR and central KOR may be involved in the modulation of scratching behaviour in imiquimod-treated mice.


Subject(s)
Aminoquinolines , Behavior, Animal , Drug Eruptions/metabolism , Psoriasis/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Skin/metabolism , Spinal Cord/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Drug Eruptions/etiology , Drug Eruptions/prevention & control , Drug Eruptions/psychology , Imiquimod , Male , Mice, Inbred C57BL , Narcotic Antagonists/pharmacology , Psoriasis/chemically induced , Psoriasis/prevention & control , Psoriasis/psychology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Signal Transduction/drug effects , Skin/drug effects , Skin/innervation , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...