1
|
DeBerg HA, Fahning ML, Varkhande SR, Schlenker JD, Schmitt WP, Gupta A, Singh A, Gratz IK, Carlin JS, Campbell DJ, Morawski PA. T Cells Promote Distinct Transcriptional Programs of Cutaneous Inflammatory Disease in Keratinocytes and Dermal Fibroblasts. J Invest Dermatol 2025:S0022-202X(25)00401-4. [PMID: 40216155 DOI: 10.1016/j.jid.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/06/2025] [Accepted: 03/23/2025] [Indexed: 04/25/2025]
Abstract
T cells and structural cells coordinate appropriate inflammatory responses and restoration of barrier integrity following insult. Dysfunctional T cells precipitate skin pathology occurring alongside altered structural cell frequencies and transcriptional states, but to what extent different T cells promote disease-associated changes remains unclear. We show that functionally diverse circulating and skin-resident CD4+CLA+ T-cell populations promote distinct transcriptional outcomes in human keratinocytes and fibroblasts associated with inflamed or healthy tissue. We identify T helper 17 cell-induced genes in keratinocytes that are enriched in psoriasis patient skin and normalized by anti-IL-17 therapy. We also describe a CD103+ skin-resident T-cell-induced transcriptional module enriched in healthy controls that is diminished during psoriasis and scleroderma and show that CD103+ T-cell frequencies are altered during disease. Interrogating clinical data using immune-dependent transcriptional signatures defines the T-cell subsets and genes distinguishing inflamed from healthy skin and allows investigation of heterogeneous patient responses to biologic therapy.
Collapse
Affiliation(s)
- Hannah A DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Mitch L Fahning
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Suraj R Varkhande
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - James D Schlenker
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, Washington, USA
| | - William P Schmitt
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Aayush Gupta
- Department of Dermatology, Leprology, and Venereology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, India
| | - Archana Singh
- Systems Biology Lab, CSIR - Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Gaziabad, India
| | - Iris K Gratz
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA; Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria; EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria; Center for Tumor Biology and Immunology, University of Salzburg, Salzburg, Austria
| | - Jeffrey S Carlin
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA; Division of Rheumatology, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Daniel J Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA; Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Peter A Morawski
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA.
| |
Collapse
|
2
|
Strazzabosco G, Liboni A, Pezzi G, Alogna A, Bortolotti D. Insights into Liposomal and Gel-Based Formulations for Dermatological Treatments. Gels 2025; 11:245. [PMID: 40277680 PMCID: PMC12027463 DOI: 10.3390/gels11040245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/26/2025] Open
Abstract
Dermatological diseases pose a significant challenge due to their chronic nature, complex pathophysiology, and the need for effective, patient-friendly treatments. Recent advancements in liposomal and gel-based formulations have played a crucial role in improving drug delivery, therapeutic efficacy, and patient compliance. Liposomal formulations have garnered considerable attention in dermatology due to their ability to encapsulate both hydrophilic and lipophilic compounds, enabling controlled drug release and enhanced skin penetration. However, challenges such as formulation complexity, stability issues, and regulatory constraints remain. Similarly, gel-based formulations are widely used due to their ease of application, biocompatibility, and ability to retain active ingredients. However, they also face limitations, including restricted penetration depth, susceptibility to microbial contamination, and challenges in achieving sustained drug release. The integration of liposomal and gel-based technologies offers a promising strategy to overcome current challenges and optimize dermatological drug delivery. This review explores both well-established therapies and recent innovations, offering a comprehensive overview of their applications in the treatment of prevalent dermatological conditions. Ultimately, continued research is essential to refine these formulations, expanding their clinical utility and enhancing therapeutic effectiveness in dermatology.
Collapse
Affiliation(s)
- Giovanni Strazzabosco
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.S.); (A.L.)
| | - Alessia Liboni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.S.); (A.L.)
| | - Giulia Pezzi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| | - Andrea Alogna
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| | - Daria Bortolotti
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| |
Collapse
|
3
|
Indolfi C, Grella C, Klain A, Dinardo G, Colosimo S, Piatto D, Nespoli C, Perrotta A, Miraglia del Giudice M. Biomarkers in Atopic Dermatitis in Children: A Comprehensive Review. Life (Basel) 2025; 15:375. [PMID: 40141720 PMCID: PMC11943560 DOI: 10.3390/life15030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder with significant implications for patient quality of life and a well-documented association with the atopic march. Recent advancements in biomarker research have unveiled critical insights into AD pathogenesis, diagnosis, prognosis, and therapeutic monitoring. This comprehensive review evaluates the utility of emerging biomarkers, including cytokines, chemokines, genetic markers, and microbiome-related components, in understanding the disease mechanisms and stratifying patient care. The role of minimally invasive diagnostic techniques, such as tape stripping and RNA monitoring, is highlighted, offering innovative approaches to pediatric populations. Furthermore, this review explores the biomarkers that predict disease progression, therapeutic response, and comorbidities, including food allergies and asthma. Personalized treatment strategies based on endotype-specific biomarkers are discussed as a future direction for improving clinical outcomes. Despite promising findings, the integration of biomarkers into routine practice necessitates further validation through large-scale studies. This work underscores the transformative potential of biomarker-driven approaches in enhancing the management of AD in children and its associated conditions.
Collapse
Affiliation(s)
| | | | - Angela Klain
- Department of Woman, Child and General and Specialized Surgery, University of Campania ‘Luigi Vanvitelli’, 80138 Naples, Italy; (C.I.); (C.G.); (S.C.); (D.P.); (C.N.); (A.P.); (M.M.d.G.)
| | - Giulio Dinardo
- Department of Woman, Child and General and Specialized Surgery, University of Campania ‘Luigi Vanvitelli’, 80138 Naples, Italy; (C.I.); (C.G.); (S.C.); (D.P.); (C.N.); (A.P.); (M.M.d.G.)
| | | | | | | | | | | |
Collapse
|
4
|
DeBerg HA, Fahning ML, Varkhande SR, Schlenker JD, Schmitt WP, Gupta A, Singh A, Gratz IK, Carlin JS, Campbell DJ, Morawski PA. T cells promote distinct transcriptional programs of cutaneous inflammatory disease in keratinocytes and dermal fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606077. [PMID: 39131334 PMCID: PMC11312529 DOI: 10.1101/2024.07.31.606077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
T cells and structural cells coordinate appropriate inflammatory responses and restoration of barrier integrity following insult. Dysfunctional T cells precipitate skin pathology occurring alongside altered structural cell frequencies and transcriptional states, but to what extent different T cells promote disease-associated changes remains unclear. We show that functionally diverse circulating and skin-resident CD4+CLA+ T cell populations promote distinct transcriptional outcomes in human keratinocytes and fibroblasts associated with inflamed or healthy tissue. We identify Th17 cell-induced genes in keratinocytes that are enriched in psoriasis patient skin and normalized by anti-IL-17 therapy. We also describe a CD103+ skin-resident T cell-induced transcriptional module enriched in healthy controls that is diminished during psoriasis and scleroderma and show that CD103+ T cell frequencies are altered during disease. Interrogating clinical data using immune-dependent transcriptional signatures defines the T cell subsets and genes distinguishing inflamed from healthy skin and allows investigation of heterogeneous patient responses to biologic therapy.
Collapse
Affiliation(s)
- Hannah A. DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Mitch L. Fahning
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Suraj R. Varkhande
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - James D. Schlenker
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, WA, USA
| | - William P. Schmitt
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, WA, USA
| | - Aayush Gupta
- Department of Dermatology, Leprology, and Venereology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pune, India
| | - Archana Singh
- Systems Biology Lab, CSIR – Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Gaziabad, India
| | - Iris K. Gratz
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
- EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- Center for Tumor Biology and Immunology, University of Salzburg, Salzburg, Austria
| | - Jeffrey S. Carlin
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
- Division of Rheumatology, Virginia Mason Medical Center, Seattle, WA, USA
| | - Daniel J. Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Peter A. Morawski
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
5
|
Liu Q, Xia Y, Liu L, Zhou Y, Li Y. Recent progress in tyrosine kinase 2 inhibitors for atopic dermatitis. Expert Opin Investig Drugs 2024; 33:1001-1007. [PMID: 39145899 DOI: 10.1080/13543784.2024.2391825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by persistent itching. Conventional treatments for AD include topical corticosteroids and calcineurin inhibitors, but there are emerging therapies targeting the JAK-TYK2 pathway that are promising for the treatment of AD. AREAS COVERED This review comprehensively explores the pathogenesis, triggers, clinical manifestations, and conventional treatment options for AD. In addition, we discuss novel therapeutic agents targeting alternative signaling pathways, with a focus on clinical trials evaluating tyrosine kinase 2 (TYK2) inhibitors, including systemic and topical agents. We also provide a detailed assessment of ICP-332 efficacy, safety, and potential adverse effects in moderate-to-severe AD. EXPERT OPINION Janus kinase inhibitors that have been recently approved have shown promise for the treatment of AD, especially for patients with severe phenotypes. Preliminary findings from randomized controlled trials suggest that TYK2 inhibitors exhibit rapid efficacy and acceptable safety in the management of AD; however, additional investigations, including long-term trials, are warranted to fully understand their efficacy and safety profile.
Collapse
Affiliation(s)
- Qi Liu
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuan Xia
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Laboratory for Regeneration Medicine, Jiangsu University, Zhenjiang, China
| | - Lin Liu
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Laboratory for Regeneration Medicine, Jiangsu University, Zhenjiang, China
| | - Yuan Zhou
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Laboratory for Regeneration Medicine, Jiangsu University, Zhenjiang, China
| | - Yumei Li
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
6
|
Gade L, Boyd BJ, Malmsten M, Heinz A. Stimuli-responsive drug delivery systems for inflammatory skin conditions. Acta Biomater 2024; 187:1-19. [PMID: 39209132 DOI: 10.1016/j.actbio.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/26/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Inflammatory skin conditions highly influence the quality of life of the patients suffering from these disorders. Symptoms include red, itchy and painful skin lesions, which are visible to the rest of the world, causing stigmatization and a significantly lower mental health of the patients. Treatment options are often unsatisfactory, as they suffer from either low patient adherence or the risk of severe side effects. Considering this, there is a need for new treatments, and notably of new ways of delivering the drugs. Stimuli-responsive drug delivery systems are able to deliver their drug cargo in response to a given stimulus and are, thus, promising for the treatment of inflammatory skin conditions. For example, the use of external stimuli such as ultraviolet light, near infrared radiation, or alteration of magnetic field enables drug release to be precisely controlled in space and time. On the other hand, internal stimuli induced by the pathological condition, including pH alteration in the skin or upregulation of reactive oxygen species or enzymes, can be utilized to create drug delivery systems that specifically target the diseased skin to achieve a better efficacy and safety. In the latter context, however, it is of key importance to match the trigger mechanism of the drug delivery system to the actual pathological features of the specific skin condition. Hence, the focus of this article is placed not only on reviewing stimuli-responsive drug delivery systems developed to treat specific inflammatory skin conditions, but also on critically evaluating their efficacy in the context of specific skin diseases. STATEMENT OF SIGNIFICANCE: Skin diseases affect one-third of the world's population, significantly lowering the quality of life of the patients, who deal with symptoms such as painful and itchy skin lesions, as well as stigmatization due to the visibility of their symptoms. Current treatments for inflammatory skin conditions are often hampered by low patient adherence or serious drug side effects. Therefore, more emphasis should be placed on developing innovative formulations that provide better efficacy and safety for patients. Stimuli-responsive drug delivery systems hold considerable promise in this regard, as they can deliver their cargo precisely where and when it is needed, reducing adverse effects and potentially offering better treatment outcomes.
Collapse
Affiliation(s)
- Luna Gade
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark
| | - Ben J Boyd
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Martin Malmsten
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark; Department of Physical Chemistry 1, Lund University, Lund, Sweden
| | - Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| |
Collapse
|
7
|
Zhao Y, Bai YP, Li LF. Association Between Systemic Immune-Inflammation Index and Psoriasis, Psoriasis Comorbidities, and All-Cause Mortality: A Study Based on NHANES. Immun Inflamm Dis 2024; 12:e70050. [PMID: 39467182 PMCID: PMC11515906 DOI: 10.1002/iid3.70050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVE The relationship between systemic immune-inflammation index (SII) and psoriasis and its prognosis is not yet clear. In this study, the correlation between SII and psoriasis, psoriasis comorbidities, and all-cause mortality was investigated based on the National Health and Nutrition Examination Survey (NHANES). METHODS The study population was derived from five NHANES cycles: 2003-2006, 2009-2014, and survival follow-up was as of December 31, 2019. The association between SII and psoriasis and its comorbidities was analyzed using weighted multivariate logistic regression models. Weighted COX regression was used to calculate hazard ratios (HRs) and the corresponding 95% confidence intervals (CIs). Restricted cubic spline, subgroup and sensitivity analyses were also used. Logarithmic conversion was performed on SII(log2SII) to reduce the impact of outliers. RESULTS A total of 21,431 participants were included in this study. As a continuous variable, log2SII was significantly associated with psoriasis in the fully adjusted model [OR = 1.20(1.04-1.39), p = .01]. log2SII remained positively associated with psoriasis after excluding participants with a history of cancer or cardiovascular disease (CVD), or non-Hispanic black participants. Among psoriasis patients, log2SII was significantly associated with metabolic syndrome (MetS) [OR = 1.68(1.19,2.38), p = .004] and all-cause mortality [HR = 1.48(1.09,1.99), p = .01]. Similar results were consistently observed when SII was analyzed as a categorical variable (in quartiles). CONCLUSION This study suggested a positive association between SII and the prevalence of psoriasis. Among psoriasis patients, SII was positively correlated with MetS and all-cause mortality.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Dermatology, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Yan Ping Bai
- Department of DermatologyChina‐Japan Friendship HospitalBeijingChina
| | - Lin Feng Li
- Department of Dermatology, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Zhu X, Wu W. The causal relationship between immune cells and atopic dermatitis: A bidirectional Mendelian randomization study. Skin Res Technol 2024; 30:e13858. [PMID: 39196303 DOI: 10.1111/srt.13858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin condition whose origins remain unclear. Existing epidemiological evidence suggests that inflammation and immune factors play pivotal roles in the onset and progression of AD. However, previous research on the connection between immune inflammation and AD has yielded inconclusive results. METHODS To evaluate the causal relationship between immunological characteristics and AD, this study employed a bidirectional, two-sample Mendelian randomization (MR) approach. We utilized large-scale, publicly available genome-wide association studies to investigate the causal associations between 731 immunological feature cells and the risk of AD. RESULTS Significant associations were identified between six immune phenotypes and AD risk: increased Basophil %CD33dim HLA DR-CD66b-, CD25 on IgD+ CD24+, CD40 on monocytes, HLA DR on CD14+ CD16-monocytes, HLA DR on CD14+monocytes correlated with higher AD risk, while elevated CD3 on CD4 Treg was linked to lower risk. Reverse MR analysis revealed AD as a risk factor for IgD+ CD38br AC and IgD+ CD38br %B cell, but a protective factor against CD20 on IgD+ CD38- naive and CD8 on NKT. CONCLUSION Our findings elucidate the intricate interplay between immune cells and AD, informing future research into AD pathophysiology and therapeutics.
Collapse
Affiliation(s)
- Xu Zhu
- Department of dermatology, Shenzhen Second People's Hospital. The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wenzhong Wu
- Department of dermatology, Shenzhen Second People's Hospital. The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Chummun Phul I, Gómez-Llonín A, Bhaw-Luximon A. From traditional medicine to nanomedicine: potential of Ginkgo biloba extracts in treating inflammatory skin diseases. RSC Med Chem 2024; 15:2643-2656. [PMID: 39149101 PMCID: PMC11324057 DOI: 10.1039/d4md00194j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/10/2024] [Indexed: 08/17/2024] Open
Abstract
The use of plant extracts as a potential cure for various conditions has moved from traditional medicine to evidence-based medicine. Skin diseases have been addressed since time immemorial using plant extracts through observational and traditional knowledge and passed on through generations. With the advent of modern techniques, the molecular mechanisms of action of plant extracts/isolates are being deciphered with more precision, and more nanomedicine-based therapies are being studied to improve their therapeutic efficacy and stability. The leaves and seeds of Ginkgo biloba (G. biloba), an ancient medicinal tree species, have been used in Chinese herbal medicine for thousands of years. G. biloba extracts have been widely studied as a neuroprotective and anti-ischaemic drug for ischaemia-reperfusion injuries in the heart, lungs, brain, kidneys, and other organs. However, the use of G. biloba can be accompanied with side effects and drug interactions. Although, there is now a growing interest for its use in skincare, the mechanisms of action of the extract are not fully understood and vital aspects of G. biloba, such as its neuroprotective and angiogenic properties contributing to the treatment of inflammatory skin diseases and skin ageing, are yet to be investigated. This review critically discusses the mechanisms of action of different constituents of G. biloba extracts linked to their potential interference in the molecular mechanisms underlying the pathogenesis of inflammatory skin diseases. In addition to its ability to act on oxidative stress, G. biloba can regulate angiogenesis through its compounds such as ginkgetin or ginkgolide K, which either inhibit aberrant angiogenesis in eczema/psoriasis or increase microcirculation during skin ageing. G. biloba may also contribute to the control of pruritus in atopic dermatitis via a neuroprotective and anti-inflammatory mechanism by suppressing JAK2/STAT3 signalling pathways. This review also highlights nanomedicine strategies to decrease the side effects and enhance the efficacy of the extracts. Similar strategies have been successfully used for anticancer molecules in targeted chemotherapy and iron delivery in anaemia treatment.
Collapse
Affiliation(s)
- Itisha Chummun Phul
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| | - Andrea Gómez-Llonín
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| | - Archana Bhaw-Luximon
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| |
Collapse
|
10
|
Shin SH, Kim JY, Kim S, Kim JH. Comparative study of efficacy and safety: Biosimilar rituximab versus originator rituximab in the treatment of pemphigus. J Dermatol 2024; 51:1104-1107. [PMID: 38874429 PMCID: PMC11483930 DOI: 10.1111/1346-8138.17329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Rituximab is a monoclonal antibody that targets CD20 antigen in B cells. For pemphigus, rituximab has been highly effective in steroid-sparing therapy for moderate to severe cases. Originator rituximab has demonstrated favorable treatment effects in patients with pemphigus, but its high cost remains a challenge. Biosimilar rituximab is expected to offer a potential solution. However, it is required for the comparative study of efficacy and safety between biosimilar and originator because all biosimilars may not be identical to the originator. In this study, we compared the treatment effects and safety of biosimilar (Truxima) and originator (MabThera) rituximab in patients with pemphigus. A final cohort of 52 patients in the MabThera group and 72 patients in the Truxima group was enrolled. Except for the intravenous immunoglobulin administration rate, there were no differences in baseline characteristics between the two groups, and for the purpose of comparing efficacy, investigations into time to complete remission, total steroid intake to complete remission, and total steroid intake for 6 months following rituximab treatment revealed no significant differences between the two groups. Truxima can be considered a relatively affordable alternative treatment option for pemphigus, offering cost-effectiveness to patients who are indicated for the treatment with MabThera.
Collapse
Affiliation(s)
- Sang Heon Shin
- Department of MedicineYonsei University College of MedicineSeoulKorea
| | - Jae Yeon Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research InstituteYonsei University College of MedicineSeoulKorea
| | - Soo‐Chan Kim
- Department of Dermatology, Yongin Severance Hospital, Cutaneous Biology Research InstituteYonsei University College of MedicineSeoulGyeonggi‐doKorea
| | - Jong Hoon Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research InstituteYonsei University College of MedicineSeoulKorea
| |
Collapse
|
11
|
Alves-Silva JM, Pedreiro S, Zuzarte M, Cruz MT, Figueirinha A, Salgueiro L. Unlocking the Bioactive Potential and Exploring Novel Applications for Portuguese Endemic Santolina impressa. PLANTS (BASEL, SWITZERLAND) 2024; 13:1943. [PMID: 39065470 PMCID: PMC11280954 DOI: 10.3390/plants13141943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
The infusion of Santolina impressa, an endemic Portuguese plant, is traditionally used to treat various infections and disorders. This study aimed to assess its chemical profile by HPLC-DAD-ESI-MSn and validate its anti-inflammatory potential. In addition, the antioxidant capacity and effects on wound healing, lipogenesis, melanogenesis, and cellular senescence, all processes in which a dysregulated inflammatory response plays a pivotal role, were unveiled. The anti-inflammatory potential was assessed in lipopolysaccharide (LPS)-stimulated macrophages, cell migration was determined using a scratch wound assay, lipogenesis was assessed on T0901317-stimulated keratinocytes and melanogenesis on 3-isobutyl-1-methylxanthine (IBMX)-activated melanocytes. Etoposide was used to induce senescence in fibroblasts. Our results point out a chemical composition predominantly characterized by dicaffeoylquinic acids and low amounts of flavonols. Regarding the infusion's bioactive potential, an anti-inflammatory effect was evident through a decrease in nitric oxide production and inducible nitric oxide synthase and pro-interleukin-1β protein levels. Moreover, a decrease in fibroblast migration was observed, as well as an inhibition in both intracellular lipid accumulation and melanogenesis. Furthermore, the infusion decreased senescence-associated β-galactosidase activity, γH2AX nuclear accumulation and both p53 and p21 protein levels. Overall, this study confirms the traditional uses of S. impressa and ascribes additional properties of interest in the pharmaceutical and dermocosmetics industries.
Collapse
Affiliation(s)
- Jorge M. Alves-Silva
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (J.M.A.-S.); (M.Z.)
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
| | - Sónia Pedreiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (J.M.A.-S.); (M.Z.)
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
| | - Maria Teresa Cruz
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra Center for Neuroscience and Cell Biology (CNC-UC), Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; (S.P.); (M.T.C.); (A.F.)
- Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, 3030-790 Coimbra, Portugal
| |
Collapse
|
12
|
Anitua E, Tierno R, Azkargorta M, Elortza F, Alkhraisat MH. Effect of Health Status and Heat-Induced Inactivation on the Proteomic Profile of Plasma Rich in Growth Factors Obtained from Donors with Chronic Inflammatory Skin Conditions. Biomolecules 2024; 14:763. [PMID: 39062477 PMCID: PMC11275043 DOI: 10.3390/biom14070763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Atopic dermatitis, psoriasis and lichen sclerosus are among the most challenging conditions treated by dermatologists worldwide, with potentially significant physical, social and psychological impacts. Emerging evidence suggests that autologous-platelet-rich plasma could be used to manage skin inflammation. However, the presence of soluble autoimmune components could hinder their therapeutic potential. The aim of this study was to analyze the proteomic profile of plasma rich in growth factors (PRGFs) obtained from donors with inflammatory skin conditions to evaluate the impact of skin health status on the composition and bioactivity of PRGF-based treatments. Venous blood from healthy volunteers and patients with psoriasis, lichen sclerosus and atopic dermatitis was processed to produce PRGF supernatant. Half of the samples were subjected to an additional thermal treatment (56 °C) to inactivate inflammatory and immune molecules. Proteomic analysis was performed to assess the protein profile of PRGFs from healthy and non-healthy patients and the effect of Immunosafe treatment. Differential abundance patterns of several proteins related to key biological processes have been identified, including complement activation, blood coagulation, and glycolysis- and gluconeogenesis-related genes. These results also demonstrate that the thermal treatment (Immunosafe) contributes to the inactivation of the complement system and, as a consequence, reduction in the immunogenic potential of PRGF products.
Collapse
Affiliation(s)
- Eduardo Anitua
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| | - Roberto Tierno
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Mohammad H. Alkhraisat
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| |
Collapse
|
13
|
Mantle D, Hargreaves IP. Coenzyme Q10 and Autoimmune Disorders: An Overview. Int J Mol Sci 2024; 25:4576. [PMID: 38674161 PMCID: PMC11049925 DOI: 10.3390/ijms25084576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/13/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
Some 90 autoimmune disorders have been described in medical literature, affecting most of the tissues within the body. Autoimmune disorders may be difficult to treat, and there is a need to develop novel therapeutic strategies for these disorders. Autoimmune disorders are characterised by mitochondrial dysfunction, oxidative stress, and inflammation; there is therefore a rationale for a role for coenzyme Q10 in the management of these disorders, on the basis of its key role in normal mitochondrial function, as an antioxidant, and as an anti-inflammatory agent. In this article, we have therefore reviewed the potential role of CoQ10, in terms of both deficiency and/or supplementation, in a range of autoimmune disorders.
Collapse
Affiliation(s)
| | - Iain P. Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| |
Collapse
|
14
|
Lee SG, Kim SC, Kim JH. Risk factors of rituximab-induced thrombocytopenia in patients with autoimmune bullous diseases. J Dermatol 2024; 51:597-601. [PMID: 37830427 DOI: 10.1111/1346-8138.17006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
Rituximab has been the mainstay treatment for autoimmune bullous diseases (AIBDs). Among the side effects of rituximab, rituximab-induced thrombocytopenia (RIT) is a rare but critical complication. However, there have been no reports or identification of risk factors for RIT in patients with AIBD. In our retrospective study, we compared rituximab-treated AIBD in patients with and without thrombocytopenia to explore the risk factors. In addition, we compared two different rituximab protocols (rheumatoid arthritis [RA] and lymphoma) in terms of the incidence and severity of thrombocytopenia. A total of 222 patients were enrolled, and 46 patients (20.7%) developed RIT. Multivariate logistic regression analysis identified age and chronic kidney disease (CKD) as significant factors for RIT. We also found that patients treated with the lymphoma protocol demonstrated a significantly higher mean post-rituximab platelet count compared with those on the RA protocol. This was the first analysis, to our knowledge, of risk factors for RIT in patients with AIBD. Individuals aged 70 or older and those with multiple comorbidities, particularly CKD, should be closely monitored for thrombocytopenia. For patients with CKD, it may be safer to use the lymphoma protocol for rituximab administration as it results in a lesser reduction in post-rituximab platelet count.
Collapse
Affiliation(s)
- Sang Gyun Lee
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Chan Kim
- Department of Dermatology, Yongin Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Yongin, Korea
| | - Jong Hoon Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Han D, Lee AY, Kim T, Choi JY, Cho MY, Song A, Kim C, Shim JH, Kim HJ, Kim H, D’Angio HB, Preska R, Mayer AT, Kim M, Choi EJ, Kim TG, Shin EC, Park K, Kim DY, Kim SC, Kim JH. Microenvironmental network of clonal CXCL13+CD4+ T cells and Tregs in pemphigus chronic blisters. J Clin Invest 2023; 133:e166357. [PMID: 37815865 PMCID: PMC10688981 DOI: 10.1172/jci166357] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUNDPemphigus, a rare autoimmune bullous disease mediated by antidesmoglein autoantibodies, can be controlled with systemic medication like rituximab and high-dose systemic corticosteroids combined with immunosuppressants. However, some patients continue to experience chronically recurrent blisters in a specific area and require long-term maintenance systemic therapy.METHODSSkin with chronic blisters was obtained from patients with pemphigus. Immunologic properties of the skin were analyzed by immunofluorescence staining, bulk and single-cell RNA and TCR sequencing, and a highly multiplex imaging technique known as CO-Detection by indEXing (CODEX). Functional analyses were performed by flow cytometry and bulk RNA-Seq using peripheral blood from healthy donors. Intralesional corticosteroid was injected into patient skin, and changes in chronically recurrent blisters were observed.RESULTSWe demonstrated the presence of skin tertiary lymphoid structures (TLSs) with desmoglein-specific B cells in chronic blisters from patients with pemphigus. In the skin TLSs, CD4+ T cells predominantly produced CXCL13. These clonally expanded CXCL13+CD4+ T cells exhibited features of activated Th1-like cells and downregulated genes associated with T cell receptor-mediated signaling. Tregs are in direct contact with CXCL13+CD4+ memory T cells and increased CXCL13 production of CD4+ T cells through IL-2 consumption and TGF-β stimulation. Finally, intralesional corticosteroid injection improved chronic blisters and reduced skin TLSs in patients with pemphigus.CONCLUSIONThrough this study we conclude that skin TLSs are associated with the persistence of chronically recurrent blisters in patients with pemphigus, and the microenvironmental network involving CXCL13+CD4+ T cells and Tregs within these structures plays an important role in CXCL13 production.TRIAL REGISTRATIONClinicalTrials.gov NCT04509570.FUNDINGThis work was supported by National Research Foundation of South Korea (NRF-2021R1C1C1007179) and Korea Drug Development Fund, which is funded by Ministry of Science and ICT; Ministry of Trade, Industry, and Energy; and Ministry of Health and Welfare (grant RS-2022-00165917).
Collapse
Affiliation(s)
- Dawoon Han
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - A Yeong Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Taehee Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Young Choi
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Mi Yeon Cho
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ahreum Song
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Changhyeon Kim
- Department of Computer Science and Engineering, College of Information and Biotechnology, National Institute of Science and Technology, Ulsan, South Korea
| | - Joon Ho Shim
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyun Je Kim
- Genome Medicine Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Honesty Kim
- Enable Medicine, Menlo Park, California, USA
| | | | - Ryan Preska
- Enable Medicine, Menlo Park, California, USA
| | | | - Miri Kim
- Yeouido St. Mary’s Hospital College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Ji Choi
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Tae-Gyun Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Kyemyung Park
- Graduate School of Health Science and Technology, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Do-Young Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Soo-Chan Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, South Korea
| | - Jong Hoon Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Shakur AA, Ranjan RK, Sinha R, Hameed S, Mohan L. A Study of Drug Utilization Pattern and Pharmacoeconomic Analysis of Immunosuppressant Drugs in Patients With Skin Disorders in a Tertiary Care Hospital in Bihar. Cureus 2023; 15:e48541. [PMID: 37954626 PMCID: PMC10632555 DOI: 10.7759/cureus.48541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
Aims The cost-effective therapy of immunosuppressant drugs in dermatological conditions will not only lead to adherence to rational prescribing but will also increase patient compliance with fewer dropouts due to cost factor. Thus, this study was done to determine and compare the drug utilization pattern, prescribed daily dose/defined daily dose (PDD/DDD) defined by WHO, and the cost-effectiveness ratio of different immunosuppressants. Methods and material Prescriptions for patients with skin disorders prescribed with any one systematic or topical immunosuppressant were collected. The utilization of drugs in different skin disorders was expressed as frequency and percentage. PDD was compared with DDD as per the WHOCC-Anatomical Therapeutic Chemical (ATC)/DDD index. The pharmacoeconomic analysis was done using a cost-effectiveness ratio. Statistical analysis Descriptive statistics were used to calculate percentages, frequency, and 95% CI. The cost-effectiveness ratio in terms of SFDs (symptom-free days) was defined as the total cost of the initial antibiotic during the study period divided by the number of SFDs (cost/ SFD) and was expressed as mean±standard deviation, and the Kruskal-Wallis test was used to determine statistical significance of difference. Results Immunosuppressants were prescribed in 117 (19.12%) prescriptions out of a total of 612 prescriptions. Deflazacort was the most utilized systemic immunosuppressant prescribed in 27.18% of cases and was commonly prescribed for irritant contact dermatitis (ICD) and allergic contact dermatitis (ACD) followed by prednisolone and betamethasone. Tacrolimus was the most utilized topical immunosuppressant prescribed in 15.90% of patients and was commonly used for ICD and vitiligo followed by clobetasol and mometasone. Betamethasone, prednisolone, clobetasol, and mometasone had better cost-effectiveness. PDD/DDD of all immunosuppressants was less than one except prednisolone, which had a PDD/DDD ratio of 3.52. Conclusions The cost-effectiveness of steroids has the advantage of providing better patients' adherence to pharmacotherapy, but over-prescribing could also lead to long-term adverse effects of steroids. Pharmacovigilance research should also incorporate pharmacoeconomic analysis to determine the relation between these two aspects.
Collapse
Affiliation(s)
- Adil A Shakur
- Department of Pharmacology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Raushan K Ranjan
- Department of Pharmacology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Rajesh Sinha
- Department of Skin and Venereal Diseases, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Saajid Hameed
- Department of Pharmacology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Lalit Mohan
- Department of Pharmacology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| |
Collapse
|
17
|
Alves-Silva JM, Moreira P, Cavaleiro C, Pereira C, Cruz MT, Salgueiro L. Effect of Ferulago lutea (Poir.) Grande Essential Oil on Molecular Hallmarks of Skin Aging. PLANTS (BASEL, SWITZERLAND) 2023; 12:3741. [PMID: 37960097 PMCID: PMC10648677 DOI: 10.3390/plants12213741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
With the increase in global life expectancy, maintaining health into old age becomes a challenge, and research has thus concentrated on various strategies which aimed to mitigate the effects of skin aging. Aromatic plants stand out as promising sources of anti-aging compounds due to their secondary metabolites, particularly essential oils (EOs). The aim of this study was to ascribe to Ferulago lutea EO several biological activities that could be useful in the context of skin aging. The EO was obtained using hydrodistillation and characterized by gas chromatography-mass spectrometry (GC/MS). The anti-inflammatory potential was assessed using lipopolysaccharide (LPS)-stimulated macrophages. The effect on cell migration was disclosed using scratch wound assay. Lipogenesis was induced using T0901317, hyperpigmentation with 3-isobutyl-1-methylxantine (IBMX) and senescence with etoposide. Our results show that the EO was characterized mainly by α-pinene and limonene. The EO was able to decrease nitric oxide (NO) release as well as iNOS and pro-IL-1β protein levels. The EO promoted wound healing while decreasing lipogenesis and having depigmenting effects. The EO also reduced senescence-associated β-galactosidase, p21/p53 protein levels and the nuclear accumulation of γH2AX. Overall, our study highlights the properties of F. lutea EO that make it a compelling candidate for dermocosmetics applications.
Collapse
Affiliation(s)
- Jorge M. Alves-Silva
- Univ Coimbra, Institute for Clinical and Biomedical Research, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
| | - Patrícia Moreira
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
| | - Carlos Cavaleiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, 3030-790 Coimbra, Portugal
| | - Cláudia Pereira
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology, 3000-548 Coimbra, Portugal; (P.M.); (C.P.)
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
- Univ Coimbra, Faculty of Medicine, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
| | - Maria Teresa Cruz
- Univ Coimbra, Center for Neuroscience and Cell Biology, Faculty of Medicine, Rua Larga, 3004-504 Coimbra, Portugal;
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, 3030-790 Coimbra, Portugal
| |
Collapse
|
18
|
Lee WS, Nam KH, Kim JH, Kim WJ, Kim JE, Shin EC, Kim GR, Choi JM. Alleviating psoriatic skin inflammation through augmentation of Treg cells via CTLA-4 signaling peptide. Front Immunol 2023; 14:1233514. [PMID: 37818377 PMCID: PMC10560854 DOI: 10.3389/fimmu.2023.1233514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by hyperplasia of keratinocytes and immune cell infiltration. The IL-17-producing T cells play a key role in psoriasis pathogenesis, while regulatory T (Treg) cells are diminished during psoriatic inflammation. Current psoriasis treatments largely focus on IL-17 and IL-23, however, few studies have explored therapeutic drugs targeting an increase of Treg cells to control immune homeostasis. In this study, we investigated the effects of a cytotoxic T lymphocyte antigen-4 (CTLA-4) signaling peptide (dNP2-ctCTLA-4) in Th17, Tc17, γδ T cells, Treg cells in vitro and a mouse model of psoriasis. Treatment with dNP2-ctCTLA-4 peptide showed a significant reduction of psoriatic skin inflammation with increased Treg cell proportion and reduced IL-17 production by T cells, indicating a potential role in modulating psoriatic skin disease. We compared dNP2-ctCTLA-4 with CTLA-4-Ig and found that only dNP2-ctCTLA-4 ameliorated the psoriasis progression, with increased Treg cells and inhibited IL-17 production from γδ T cells. In vitro experiments using a T cell-antigen presenting cell co-culture system demonstrated the distinct mechanisms of dNP2-ctCTLA-4 compared to CTLA-4-Ig in the induction of Treg cells. These findings highlight the therapeutic potential of dNP2-ctCTLA-4 peptide in psoriasis by augmenting Treg/Teff ratio, offering a new approach to modulating the disease.
Collapse
Affiliation(s)
- Woo-Sung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Ho Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jong Hoon Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won-Ju Kim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Jeong Eun Kim
- Department of Dermatology, Hanyang University College of Medicine, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Gil-Ran Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Marques MP, Varela C, Mendonça L, Cabral C. Nanotechnology-Based Topical Delivery of Natural Products for the Management of Atopic Dermatitis. Pharmaceutics 2023; 15:1724. [PMID: 37376172 DOI: 10.3390/pharmaceutics15061724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic eczematous inflammatory disease that may arise from environmental, genetic, and immunological factors. Despite the efficacy of current treatment options such as corticosteroids, such approaches are mainly focused on symptom relief and may present certain undesirable side effects. In recent years, isolated natural compounds, oils, mixtures, and/or extracts have gained scientific attention because of their high efficiency and moderate to low toxicity. Despite their promising therapeutic effects, the applicability of such natural healthcare solutions is somewhat limited by their instability, poor solubility, and low bioavailability. Therefore, novel nanoformulation-based systems have been designed to overcome these limitations, thus enhancing the therapeutic potential, by promoting the capacity of these natural drugs to properly exert their action in AD-like skin lesions. To the best of our knowledge, this is the first literature review that has focused on summarizing recent nanoformulation-based solutions loaded with natural ingredients, specifically for the management of AD. We suggest that future studies should focus on robust clinical trials that may confirm the safety and effectiveness of such natural-based nanosystems, thus paving the way for more reliable AD treatments.
Collapse
Affiliation(s)
- Mário Pedro Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carla Varela
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Chemical Process Engineering and Forest Products (CIEPQPF), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Laura Mendonça
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Célia Cabral
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
20
|
Politi FAS, Carvalho SG, Rodero CF, Dos Santos KP, Meneguin AB, Sorrechia R, Chiavacci LA, Chorilli M. Piperine-loaded nanoparticles incorporated into hyaluronic acid/sodium alginate-based membranes for the treatment of inflammatory skin diseases. Int J Biol Macromol 2023; 227:736-748. [PMID: 36549615 DOI: 10.1016/j.ijbiomac.2022.12.147] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Piperine is an alkaloid mostly found in the fruits of several species of the Piper genus, and its anti-inflammatory potential is already known. However, its therapeutic applications still need to be better explored due to the low aqueous solubility of this active. To overcome this drawback, the objective of this work was to evaluate the efficiency of the nanoencapsulation of the compound as well as its incorporation into hyaluronic acid/alginate-based biomembranes. Polymeric nanoparticles composed of Eudragit S100 and Poloxamer 188 were obtained by the nanoprecipitation technique, obtaining spherical nanosized particles with an average diameter of 122.1 ± 2.0 nm, polydispersity index of 0.266, and encapsulation efficiency of 76.2 %. Hyaluronic acid/sodium alginate membranes were then prepared and characterized. Regarding permeation, a slow passage rate was observed until the initial 14 h, when an exponential increase in the recovered drug concentration began to occur. The in vivo assay showed a reduction in inflammation up to 43.6 %, and no cytotoxicity was observed. The results suggested the potential of the system developed for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Flávio Augusto Sanches Politi
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Suzana Gonçalves Carvalho
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Camila Fernanda Rodero
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Kaio Pini Dos Santos
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Andréia Bagliotti Meneguin
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Rodrigo Sorrechia
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Leila Aparecida Chiavacci
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| |
Collapse
|
21
|
Lee AY, Kim T, Kim JH. Understanding CD4 + T cells in autoimmune bullous diseases. Front Immunol 2023; 14:1161927. [PMID: 37138879 PMCID: PMC10149917 DOI: 10.3389/fimmu.2023.1161927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Autoimmune bullous diseases (AIBDs) are a group of life-threatening blistering diseases caused by autoantibodies that target proteins in the skin and mucosa. Autoantibodies are the most important mediator in the pathogenesis of AIBDs, and various immune mechanisms contribute to the production of these pathogenic autoantibodies. Recently, significant progress has been made in understanding how CD4+ T cells drive autoantibody production in these diseases. Here, we review the critical role of CD4+ T cells in the production of pathogenic autoantibodies for the initiation and perpetuation of humoral response in AIBDs. To gain an in-depth understanding of CD4+ T-cell pathogenicity, antigen specificity, and mechanisms of immune tolerance, this review covers comprehensive mouse and human studies of pemphigus and bullous pemphigoid. Further exploration of pathogenic CD4+ T cells will potentially provide immune targets for improved treatment of AIBDs.
Collapse
|
22
|
Song A, Jang J, Lee A, Min SY, Lee SG, Kim SC, Shin J, Kim JH. Clinical impact and a prognostic marker of early rituximab treatment after rituximab reimbursement in Korean pemphigus patients. Front Immunol 2022; 13:932909. [PMID: 35983042 PMCID: PMC9379325 DOI: 10.3389/fimmu.2022.932909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Pemphigus is an autoimmune mucocutaneous blistering disease caused by autoantibodies against desmogleins. Rituximab effectively treats pemphigus by inducing remission and rapidly reducing corticosteroid dosage. In Korea, the high cost of rituximab had been a burden until the National Health Insurance began to cover 90% of rituximab costs via reimbursement for severe pemphigus patients. We analyzed 214 patients with pemphigus who were treated with their first round of rituximab. The time to initiate rituximab and the time to partial remission under minimal therapy (PRMT) were both significantly shorter after the rituximab reimbursement policy. The total steroid intake for PRMT and complete remission (CR) was less in patients who were diagnosed after the reimbursement. The interrupted time series (ITS) model, a novel analysis method to evaluate the effects of an intervention, showed a decrease in total systemic corticosteroid intake until PRMT after reimbursement began. In peripheral blood mononuclear cells from patients with pemphigus vulgaris, the relative frequencies of desmoglein 3-specific CD11c+CD27−IgD− atypical memory B cells positively correlated with the periods from disease onset to rituximab treatment and to PRMT and the total systemic corticosteroid intake until PRMT. We found that early rituximab therapy, induced by the reimbursement policy, shortened the disease course and reduced the total corticosteroid use by pemphigus patients. The decreased frequency of circulating desmoglein-specific atypical memory B cells can be used as a surrogate marker for a good prognosis after rituximab.
Collapse
Affiliation(s)
- Ahreum Song
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jieun Jang
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, MA, United States
- Department of Hospital Administration, Yonsei University Graduate School of Public Health, Seoul, South Korea
| | - Ayeong Lee
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seo Yeon Min
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Gyun Lee
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Soo-Chan Kim
- Department of Dermatology, Yongin Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaeyong Shin
- Department of Preventive Medicine and Institute of Health Services Research, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jaeyong Shin, ; Jong Hoon Kim,
| | - Jong Hoon Kim
- Department of Dermatology, Gangnam Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jaeyong Shin, ; Jong Hoon Kim,
| |
Collapse
|
23
|
Stănescu AMA, Cristea AMA, Bejan GC, Vieru M, Simionescu AA, Popescu FD. Allergic Contact Cell-Mediated Hypersensitivity in Psoriasis: A Narrative Minireview. Medicina (B Aires) 2022; 58:914. [PMID: 35888633 PMCID: PMC9324524 DOI: 10.3390/medicina58070914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
The dysfunctionality of the protective skin barrier in psoriasis allows easier cutaneous penetration of various contact haptens; thus, such patients can develop allergic contact hypersensitivity as a comorbidity. Both skin conditions involve T-cell-mediated mechanisms. Dermatologists and allergists should consider assessing allergic contact cell-mediated hypersensitivity in selected psoriasis patients, especially those with palmoplantar psoriasis and who are refractory to topical treatments, and in patients with psoriasis, with or without arthritis, treated with biologics that present skin lesions clinically suggestive of contact dermatitis.
Collapse
Affiliation(s)
| | - Ana-Maria-Antoaneta Cristea
- Department of Allergology and Clinical Immunology, Nicolae Malaxa Clinical Hospital, 022441 Bucharest, Romania; (A.-M.-A.C.); (F.-D.P.)
| | - Gabriel Cristian Bejan
- Department of Family Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mariana Vieru
- Department of Allergology and Clinical Immunology, Nicolae Malaxa Clinical Hospital, 022441 Bucharest, Romania; (A.-M.-A.C.); (F.-D.P.)
- Department of Allergology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Anca Angela Simionescu
- Department of Obstetrics and Gynecology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 011132 Bucharest, Romania
| | - Florin-Dan Popescu
- Department of Allergology and Clinical Immunology, Nicolae Malaxa Clinical Hospital, 022441 Bucharest, Romania; (A.-M.-A.C.); (F.-D.P.)
- Department of Allergology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|