1
|
Lee MY, Han K, Min KH, Yu DS, Lee YB. Psoriasis as a Predictor of Neovascular Age-Related Macular Degeneration in Type 2 Diabetes Mellitus: A Nationwide Cohort Study. Am J Ophthalmol 2025; 269:236-245. [PMID: 39218384 DOI: 10.1016/j.ajo.2024.08.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The objective of this study is to evaluate the relationship of psoriasis and neovascular age-related macular degeneration (nAMD) in a population with diabetic mellitus (DM). DESIGN Nationwide, population-based, retrospective cohort study. METHODS Records of patients over 40 years of age who had been diagnosed with type 2 diabetes mellitus from January 2009 to December 2012 were analyzed. The incidence of nAMD was observed from the index year to December 2018 in all subjects. The incidence rate of nAMD was compared between the psoriasis group and control group. Covariates include age, sex, body mass index, income level, smoking status, drinking status, regular exercise habits, hypertension, dyslipidemia, end-stage renal disease, diabetic retinopathy, glucose level, prescription of more than 3 oral hypoglycemic agents, and a history of diabetes mellitus exceeding 5 years. RESULTS Of 2,245,358 patients with type 2 DM, 20,853 patients were classified in the psoriasis group and the other 2,224,505 in the control group. A total of 105 nAMD cases were observed in the psoriasis group and 7459 cases in the control group. According to multivariable Cox proportional hazard models, individuals with psoriasis had a significantly higher risk for nAMD compared to controls (hazard ratio = 1.329, 95% CI = 1.096-1.612) after adjustments for covariates. CONCLUSIONS This study demonstrated that psoriasis was an independent risk factor for developing nAMD in DM patients. Therefore, physicians should be alert to the development of nAMD in DM patients who also have psoriasis.
Collapse
Affiliation(s)
- Mee Yon Lee
- From the Department of Ophthalmology (M.Y.L.), College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science (K.H.), College of Natural Sciences, Soongsil University, Republic of Korea
| | - Kyung Hyun Min
- Department of Dermatology (K.H.M., D.S.Y., Y.B.L.), College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Dong Soo Yu
- Department of Dermatology (K.H.M., D.S.Y., Y.B.L.), College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Young Bok Lee
- Department of Dermatology (K.H.M., D.S.Y., Y.B.L.), College of Medicine, The Catholic University of Korea, Republic of Korea.
| |
Collapse
|
2
|
Mangoni AA, Zinellu A. A systematic review and meta-analysis of the endothelial-immune candidate biomarker endoglin in rheumatic diseases. Clin Exp Med 2024; 25:4. [PMID: 39535678 PMCID: PMC11561007 DOI: 10.1007/s10238-024-01519-5] [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: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Existing challenges in accurately diagnosing various rheumatic diseases (RDs) have stimulated the search for novel biomarkers to aid clinical evaluation and monitoring. We conducted a systematic review and meta-analysis of studies investigating the candidate biomarker endoglin (CD105), a transmembrane glycoprotein expressed in endothelial, myeloid, and lymphoid cells, in RD patients and healthy controls. We searched PubMed, Scopus, and Web of Science from inception to 10 August 2024 to identify relevant studies. We evaluated the risk of bias using the JBI Critical Appraisal Checklist and the certainty of evidence using GRADE (PROSPERO registration number: CRD42023581008). Overall, circulating endoglin concentrations were significantly higher in RD patients compared to controls (13 studies; standard mean difference, SMD = 0.64, 95% CI 0.13 to 1.14, p = 0.014; low certainty of evidence). The effect size of the between-group differences in endoglin concentrations was not significantly associated with age, male-to-female ratio, year of publication, number of participants, or mean RD duration. By contrast, the effect size was statistically significant in studies conducted in the European region (p = 0.033), involving patients with systemic sclerosis (p = 0.032), and measuring serum (p = 0.019). The results of this systematic review and meta-analysis suggest the potential pathophysiological role of endoglin in RDs. This, however, requires further investigation in prospective studies, particularly in patients with systemic sclerosis.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, and Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
3
|
Ebrahimi A, Mehrabi M, Miraghaee SS, Mohammadi P, Fatehi Kafash F, Delfani M, Khodarahmi R. Flavonoid compounds and their synergistic effects: Promising approaches for the prevention and treatment of psoriasis with emphasis on keratinocytes - A systematic and mechanistic review. Int Immunopharmacol 2024; 138:112561. [PMID: 38941673 DOI: 10.1016/j.intimp.2024.112561] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Psoriasis, a chronic autoimmune skin disorder, causes rapid and excessive skin cell growth due to immune system dysfunction. Numerous studies have shown that flavonoids have anti-psoriatic effects by modulating various molecular mechanisms involved in inflammation, cytokine production, keratinocyte proliferation, and more. This study reviewed experimental data reported in scientific literature and used network analysis to identify the potential biological roles of flavonoids' targets in treating psoriasis. 947 records from Web of Sciences, ScienceDirect database, Scopus, PubMed, and Cochrane library were reviewed without limitations until June 26, 2023. 66 articles were included in the systematic review. The ten genes with the highest scores, including interleukin (IL)-10, IL-12A, IL-1β, IL-6, Tumor necrosis factor-α (TNF-α), Janus kinase 2 (JAK 2), Jun N-terminal kinase (JUN), Proto-oncogene tyrosine-protein kinase Src (SRC), Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), and Signal transducer and activator of transcription 3 (STAT3), were identified as the hub genes. KEGG pathway analysis identified connections related to inflammation and autoimmune responses, which are key characteristics of psoriasis. IL-6, STAT3, and JUN's presence in both hub and enrichment genes suggests their important role in flavonoid's effect on psoriasis. This comprehensive study highlights how flavonoids can target biological processes in psoriasis, especially when combined for enhanced effectiveness.
Collapse
Affiliation(s)
- Ali Ebrahimi
- Department of Dermatology, Hajdaie Dermatology Clinic, School of Medicine, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Masomeh Mehrabi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyyed Shahram Miraghaee
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Fatehi Kafash
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohana Delfani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
4
|
Asai C, Takamura N, Watanabe T, Asami M, Ikeda N, Reese CF, Hoffman S, Yamaguchi Y. A water-soluble caveolin-1 peptide inhibits psoriasis-like skin inflammation by suppressing cytokine production and angiogenesis. Sci Rep 2024; 14:20553. [PMID: 39232048 PMCID: PMC11375059 DOI: 10.1038/s41598-024-71350-1] [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: 04/06/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
The plasma membrane protein caveolin-1 (CAV-1) regulates signaling by inhibiting a wide range of kinases and other enzymes. Our previous study demonstrated that the downregulation of CAV-1 in psoriatic epidermal cells contributes to inflammation by enhancing JAK/STAT signaling, cell proliferation, and chemokine production. Administration of the CAV-1 scaffolding domain (CSD) peptide suppressed imiquimod (IMQ)-induced psoriasis-like dermatitis. To identify an optimal therapeutic peptide derived from CAV-1, we have compared the efficacy of CSD and subregions of CSD that have been modified to make them water soluble. We refer to these modified peptides as sCSD, sA, sB, and sC. In IMQ-induced psoriasis-like dermatitis, while all four peptides showed major beneficial effects, sB caused the most significant improvements of skin phenotype and number of infiltrating cells, comparable or superior to the effects of sCSD. Phosphorylation of STAT3 was also inhibited by sB. Furthermore, sB suppressed angiogenesis both in vivo in the dermis of IMQ-induced psoriasis mice and in vitro by blocking the ability of conditioned media derived from CAV-1-silenced keratinocytes to inhibit tube formation by HUVEC. In conclusion, sB had similar or greater beneficial effects than sCSD not only by cytokine suppression but by angiogenesis inhibition adding to its ability to target psoriatic inflammation.
Collapse
Affiliation(s)
- Chika Asai
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Naoko Takamura
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Tomoya Watanabe
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Miho Asami
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Noriko Ikeda
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Charles F Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
5
|
Coto-Segura P, Segú-Vergés C, Martorell A, Moreno-Ramírez D, Jorba G, Junet V, Guerri F, Daura X, Oliva B, Cara C, Suárez-Magdalena O, Abraham S, Mas JM. A quantitative systems pharmacology model for certolizumab pegol treatment in moderate-to-severe psoriasis. Front Immunol 2023; 14:1212981. [PMID: 37809085 PMCID: PMC10552644 DOI: 10.3389/fimmu.2023.1212981] [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: 04/27/2023] [Accepted: 08/07/2023] [Indexed: 10/10/2023] Open
Abstract
Background Psoriasis is a chronic immune-mediated inflammatory systemic disease with skin manifestations characterized by erythematous, scaly, itchy and/or painful plaques resulting from hyperproliferation of keratinocytes. Certolizumab pegol [CZP], a PEGylated antigen binding fragment of a humanized monoclonal antibody against TNF-alpha, is approved for the treatment of moderate-to-severe plaque psoriasis. Patients with psoriasis present clinical and molecular variability, affecting response to treatment. Herein, we utilized an in silico approach to model the effects of CZP in a virtual population (vPop) with moderate-to-severe psoriasis. Our proof-of-concept study aims to assess the performance of our model in generating a vPop and defining CZP response variability based on patient profiles. Methods We built a quantitative systems pharmacology (QSP) model of a clinical trial-like vPop with moderate-to-severe psoriasis treated with two dosing schemes of CZP (200 mg and 400 mg, both every two weeks for 16 weeks, starting with a loading dose of CZP 400 mg at weeks 0, 2, and 4). We applied different modelling approaches: (i) an algorithm to generate vPop according to reference population values and comorbidity frequencies in real-world populations; (ii) physiologically based pharmacokinetic (PBPK) models of CZP dosing schemes in each virtual patient; and (iii) systems biology-based models of the mechanism of action (MoA) of the drug. Results The combination of our different modelling approaches yielded a vPop distribution and a PBPK model that aligned with existing literature. Our systems biology and QSP models reproduced known biological and clinical activity, presenting outcomes correlating with clinical efficacy measures. We identified distinct clusters of virtual patients based on their psoriasis-related protein predicted activity when treated with CZP, which could help unravel differences in drug efficacy in diverse subpopulations. Moreover, our models revealed clusters of MoA solutions irrespective of the dosing regimen employed. Conclusion Our study provided patient specific QSP models that reproduced clinical and molecular efficacy features, supporting the use of computational methods as modelling strategy to explore drug response variability. This might shed light on the differences in drug efficacy in diverse subpopulations, especially useful in complex diseases such as psoriasis, through the generation of mechanistically based hypotheses.
Collapse
Affiliation(s)
- Pablo Coto-Segura
- Dermatology Department, Hospital Vital Alvarez-Buylla de Mieres, Asturias, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech SL, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - David Moreno-Ramírez
- Dermatology Department, University Hospital Virgen Macarena, Andalusian Health Service, University of Seville, Seville, Spain
| | - Guillem Jorba
- Anaxomics Biotech SL, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Valentin Junet
- Anaxomics Biotech SL, Barcelona, Spain
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Filippo Guerri
- Anaxomics Biotech SL, Barcelona, Spain
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Xavier Daura
- Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Cerdanyola del Vallès, Spain
| | - Baldomero Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | - Sonya Abraham
- National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College, London, United Kingdom
- Medical Affairs, UCB Pharma, Brussels, Belgium
| | | |
Collapse
|
6
|
Niu X, Han Q, Li X, Li J, Liu Y, Li Y, Wu Y, Zhang K. EDIL3 influenced the αvβ3-FAK/MEK/ERK axis of endothelial cells in psoriasis. J Cell Mol Med 2022; 26:5202-5212. [PMID: 36065978 PMCID: PMC9575107 DOI: 10.1111/jcmm.17544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/27/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
One of the earliest events in the development of psoriatic lesion is a vascular network expansion. The abnormal vascular network is associated with increased endothelial cells (ECs) survival, proliferation, adhesion, migration, angiogenesis and permeability in psoriatic lesion. Our previous study demonstrated that epidermal growth factor‐like repeats and discoidin I‐like domains 3 (EDIL3) derived from psoriatic dermal mesenchymal stem cells (DMSCs) promoted cell–cell adhesion, migration and angiogenesis of ECs, but the molecular mechanism of upstream or downstream has not been explored. So, this study aimed to explore the association between EDIL3 derived from DMSCs (DMSCs‐derived EDIL3) and psoriasis‐associated angiogenesis. We injected recombinant EDIL3 protein to mouse model of psoriasis to confirm the roles of EDIL3 in psoriasis. Besides, we employed both short‐interference RNA (si‐RNA) and lentiviral vectors to explore the molecular mechanism of EDIL3 promoting angiogenesis in psoriasis. In vivo, this research found that after injected recombination EDIL3 protein, the epidermis thickness and microvessel density were both elevated. EDIL3 accelerated the process of psoriasis in the IMQ‐induced psoriasis‐like mouse model. Additionally, we confirmed that in vitro DMSCs‐derived EDIL3 is involved in the tube formation of ECs via αvβ3‐FAK/MEK/ERK signal pathway. This suggested that DMSCs‐derived EDIL3 and αvβ3‐FAK/MEK/ERK signal pathway in ECs play an important role in the pathogenesis of psoriasis. And the modification of DMSCs, EDIL3 and αvβ3‐FAK/MEK/ERK signal pathway will provide a valuable therapeutic target to control the angiogenesis in psoriasis.
Collapse
Affiliation(s)
- Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qixin Han
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinhua Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanmin Liu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Li
- No. 1 English Department, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Yan Wu
- Department of Dermatology, Key Laboratory of Immunodermatology, Ministry of Education and NHC, National joint Engineering Research Center for Theranostics of Immunological Skin Diseases, The First Hospital of China Medical University, Shenyang, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
7
|
Karas A, Holmannova D, Borsky P, Fiala Z, Andrys C, Hamakova K, Svadlakova T, Palicka V, Krejsek J, Rehacek V, Esterkova M, Kovarikova H, Borska L. Significantly Altered Serum Levels of NAD, AGE, RAGE, CRP, and Elastin as Potential Biomarkers of Psoriasis and Aging—A Case-Control Study. Biomedicines 2022; 10:biomedicines10051133. [PMID: 35625870 PMCID: PMC9138308 DOI: 10.3390/biomedicines10051133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 01/11/2023] Open
Abstract
Background: This study aims to investigate potential markers of psoriasis and aging, and to elucidate possible connections between these two processes. Methods: The serum samples of 60 psoriatic patients and 100 controls were analysed, and the levels of four selected parameters (AGEs, RAGE, NAD, and elastin) were determined using commercial ELISA kits. Serum C-reactive protein was assayed using an immune-nephelometry method. Findings: Among the patients, the levels of CRP, AGEs, and RAGE were all increased, while the levels of NAD were reduced when compared to the control group. A negative correlation between the levels of AGEs and NAD was found. A negative correlation between age and the NAD levels among the control group was observed, however among the patients the relationship was diminished. While there was no difference in the levels of native elastin between the patients and the controls, a positive correlation between the levels of native elastin and age and a negative correlation between the levels of native elastin and the severity of psoriasis were found. Conclusions: The results of our study support the notion of psoriasis and possibly other immune-mediated diseases accelerating the aging process through sustained systemic damage. The serum levels of CRP, NAD, AGEs, and RAGE appear to be promising potential biomarkers of psoriasis. The decrease in the serum levels of NAD is associated with (pro)inflammatory states. Our analysis indicates that the levels of native elastin might strongly reflect both the severity of psoriasis and the aging process.
Collapse
Affiliation(s)
- Adam Karas
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
| | - Drahomira Holmannova
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
| | - Pavel Borsky
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
- Correspondence: ; Tel.: +420-495-816-386
| | - Zdenek Fiala
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
| | - Ctirad Andrys
- Institute of Clinical Immunology and Allergology, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Kvetoslava Hamakova
- Clinic of Dermal and Venereal Diseases, University Hospital, 500 03 Hradec Kralove, Czech Republic;
| | - Tereza Svadlakova
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
- Institute of Clinical Immunology and Allergology, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostics, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (V.P.); (H.K.)
| | - Jan Krejsek
- Institute of Clinical Immunology and Allergology, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (C.A.); (J.K.)
| | - Vit Rehacek
- Transfusion Center, University Hospital, 500 03 Hradec Kralove, Czech Republic;
| | - Monika Esterkova
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
| | - Helena Kovarikova
- Institute of Clinical Biochemistry and Diagnostics, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (V.P.); (H.K.)
| | - Lenka Borska
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (A.K.); (D.H.); (Z.F.); (T.S.); (M.E.); (L.B.)
| |
Collapse
|
8
|
Wang J, Si R, Zhang Q, Pan X, Zhang J. Tumor-microenvironmental-response Bi-functional molecules for efficient imaging and anti-tumor activity therapy. Eur J Med Chem 2022; 230:114120. [PMID: 35051748 DOI: 10.1016/j.ejmech.2022.114120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/14/2021] [Accepted: 01/09/2022] [Indexed: 12/13/2022]
Abstract
To improve the visualization and potency of anticancer agents, the diagnosis and treatment integration bi-functional molecules were constructed based on active candidate BD7, approved drug Linifanib, and monoclonal antibody Bevacizumab. Commercial available Rhodamine B was inducted to realize imaging-aided diagnosis and target efficiency monitoring for cancer cells. In order to maintain the anticancer activity of drugs, disulfide bond was incorporated as releasable group based on tumor microenviroment. After design, synthesis and structure characterization of title compounds, various biological evaluation and cancer cell imaging analysis were carried out. The results indicated that these title diagnosis and treatment integration bi-functional molecules exhibited comparable potency with that of corresponding parent drug. Meanwhile, these agents afforded good performance in cell imaging and could be used to differentiate cancer cells from normal ovarian cells in real time. Further optimization of these bi-functional molecules is ongoing to improve the potency and precision and will be reported in due course. Our findings are expected to achieve efficient screening and real-time prognostic monitoring under the premise of high anti-tumor activity for clinical application.
Collapse
Affiliation(s)
- Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Ru Si
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China.
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
9
|
Bardehle F, Sies K, Enk A, Rosenberger A, Fink C, Haenssle H. Mikrovaskuläre Pathologien bei Patienten mit Psoriasis vulgaris mittels Nagelfalzkapillarmikroskopie identifiziert: Ergebnisse einer prospektiven kontrollierten Studie. J Dtsch Dermatol Ges 2021; 19:1736-1745. [PMID: 34894181 DOI: 10.1111/ddg.14606_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/20/2021] [Indexed: 11/30/2022]
Abstract
Hintergrund: Die Psoriasis gilt als unabhängiger kardiovaskulärer Risikofaktor und Treiber einer Atherogenese. Mikrovaskuläre Veränderungen in psoriatischen Plaques sind gut beschrieben, wohingegen Veränderungen außerhalb betroffener Hautareale kaum untersucht wurden. In dieser Studie wurden Nagelfalzkapillaren von Psoriasispatienten in nicht betroffener Haut systematisch untersucht. Patienten und Methodik: Prospektive Studie mit Untersuchung von Nagelfalzkapillaren bei Psoriasispatienten im Vergleich zu gesunden Kontrollen mittels digitaler Videokapillarmikroskopie. Es wurden 21 kapillarmikroskopische Parameter bewertet und die Ergebnisse mit Charakteristika der Patienten und der Psoriasiserkrankung, mit Laborparametern und Messungen der Intima-Media-Dicke der Arteria carotis communis korreliert. Ergebnisse: Die 77 Psoriasispatienten (24 mit zusätzlicher Psoriasisarthritis) und 71 Kontrollen zeigten sich hinsichtlich demographischer Merkmale und relevanter Einflussfaktoren für eine Mikroangiopathie ausbalanciert. Im Vergleich zur Kontrollgruppe zeigten Psoriasispatienten eine signifikante Minderung der kapillaren Dichte, häufigere Kapillarerweiterung mit mehr Verzweigungen, Torquierungen und kapillaren Unregelmäßigkeiten. Zusätzlich zeigten Psoriasispatienten signifikant höhere inflammatorische Serummarker und eine gesteigerte Intima-Media-Dicke. In unserem Kollektiv bestand kein Zusammenhang zwischen Krankheitsdauer oder Schweregrad der Psoriasis und spezifischen Kapillarveränderungen. Schlussfolgerungen: Die Nagelfalzkapillaren der untersuchten Psoriasispatienten zeigten ausgeprägte mikrovaskuläre Veränderungen, welche mit erhöhten Markern einer systemischen Entzündung und Frühzeichen einer Atherosklerose korrelierten. Weitere Studien sind erforderlich, um die Rolle der digitalen Videokapillarmikroskopie in der Bewertung des kardiovaskulären Risikos von Psoriasispatienten zu untersuchen.
Collapse
|
10
|
Bardehle F, Sies K, Enk A, Rosenberger A, Fink C, Haenssle H. Nailfold videocapillaroscopy identifies microvascular pathologies in psoriasis vulgaris: Results of a prospective controlled study. J Dtsch Dermatol Ges 2021; 19:1736-1744. [PMID: 34792866 DOI: 10.1111/ddg.14606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/20/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Psoriasis is considered an independent cardiovascular risk factor, evidentially driving atherosclerosis. However, little is known about changes in the microvasculature of non-lesional skin in psoriasis patients. This study systematically examined capillary pathologies in psoriasis patients by digital video nailfold capillaroscopy. PATIENTS AND METHODS Prospective study comparing nailfold capillaries of psoriasis patients with those of healthy controls. Nailfold capillaries were evaluated for 21 parameters and results were correlated with characteristics of patients and psoriatic disease, laboratory parameters, and measurements of carotid intima-media thickness. RESULTS 77 psoriasis patients (24 patients with additional psoriatic arthritis) and 71 controls were well-matched for demographic features and for relevant confounding factors causing microangiopathy. In comparison with controls, psoriasis patients showed a significant loss of capillaries, capillary expansion with increased ramifications and tortuosity and capillary irregularities. Moreover, in psoriasis patients we found significantly elevated serum markers of inflammation and significantly increased intima-media-thickness measurements. We found no effect of disease duration nor disease activity on capillary changes. CONCLUSIONS Nailfold capillaries of psoriasis patients showed marked microvascular abnormalities accompanied by increased markers of systemic inflammation and atherosclerosis. Prospective cohort studies are needed to assess the role of nailfold capillaroscopy for predicting the cardiovascular risk of psoriasis patients.
Collapse
Affiliation(s)
| | - Katharina Sies
- Department of Dermatology, University of Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology, University of Heidelberg, Germany
| | | | - Christine Fink
- Department of Dermatology, University of Heidelberg, Germany
| | - Holger Haenssle
- Department of Dermatology, University of Heidelberg, Germany
| |
Collapse
|
11
|
Maifeld A, Wild J, Karlsen TV, Rakova N, Wistorf E, Linz P, Jung R, Birukov A, Gimenez-Rivera VA, Wilck N, Bartolomaeus T, Dechend R, Kleinewietfeld M, Forslund SK, Krause A, Kokolakis G, Philipp S, Clausen BE, Brand A, Waisman A, Kurschus FC, Wegner J, Schultheis M, Luft FC, Boschmann M, Kelm M, Wiig H, Kuehne T, Müller DN, Karbach S, Markó L. Skin Sodium Accumulates in Psoriasis and Reflects Disease Severity. J Invest Dermatol 2021; 142:166-178.e8. [PMID: 34237339 DOI: 10.1016/j.jid.2021.06.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 05/14/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022]
Abstract
Sodium can accumulate in the skin at concentrations exceeding serum levels. A high sodium environment can lead to pathogenic T helper 17 cell expansion. Psoriasis is a chronic inflammatory skin disease in which IL-17‒producing T helper 17 cells play a crucial role. In an observational study, we measured skin sodium content in patients with psoriasis and in age-matched healthy controls by Sodium-23 magnetic resonance imaging. Patients with PASI > 5 showed significantly higher sodium and water content in the skin but not in other tissues than those with lower PASI or healthy controls. Skin sodium concentrations measured by Sodium-23 spectroscopy or by atomic absorption spectrometry in ashed-skin biopsies verified the findings with Sodium-23 magnetic resonance imaging. In vitro T helper 17 cell differentiation of naive CD4+ cells from patients with psoriasis markedly induced IL-17A expression under increased sodium chloride concentrations. The imiquimod-induced psoriasis mouse model replicated the human findings. Extracellular tracer Chromium-51-EDTA measurements in imiquimod- and sham-treated skin showed similar extracellular volumes, rendering excessive water of intracellular origin. Chronic genetic IL-17A‒driven psoriasis mouse models underlined the role of IL-17A in dermal sodium accumulation and inflammation. Our data describe skin sodium as a pathophysiological feature of psoriasis, which could open new avenues for its treatment.
Collapse
Affiliation(s)
- András Maifeld
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes Wild
- Center of Cardiology - Cardiology I, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Natalia Rakova
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Elisa Wistorf
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Peter Linz
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Rebecca Jung
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Birukov
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | - Nicola Wilck
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Nephrology and Internal Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Theda Bartolomaeus
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Helios Clinic Berlin-Buch, Berlin, Germany
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University Campus Diepenbeek, Hasselt, Belgium
| | - Sofia K Forslund
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Krause
- Medical Centre for Rheumatology and Clinical Immunology, Immanuel Krankenhaus Berlin, Berlin, Germany
| | - Georgios Kokolakis
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sandra Philipp
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Björn E Clausen
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Brand
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ari Waisman
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Florian C Kurschus
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joanna Wegner
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael Schultheis
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael Boschmann
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcus Kelm
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Congenital Heart Disease, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Titus Kuehne
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Congenital Heart Disease, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Berlin, Germany
| | - Susanne Karbach
- Center of Cardiology - Cardiology I, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lajos Markó
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany; Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Lu Y, Yang Y, Zhang J, Zhang H, Ma C, Tang X, Wu J, Li L, Wei J, Chen H, Lu C, Han L. Anti-Angiogenic Efficacy of PSORI-CM02 and the Associated Mechanism in Psoriasis In Vitro and In Vivo. Front Immunol 2021; 12:649591. [PMID: 33995368 PMCID: PMC8119787 DOI: 10.3389/fimmu.2021.649591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis is a chronic proliferative autoimmune dermatologic disease characterised by abnormal angiogenesis. Thus, regulating angiogenesis in the skin is an important treatment strategy for psoriasis. PSORI-CM02, an empirical Chinese medicine formula optimised from Yin Xie Ling, was created by the Chinese medicine specialist, Guo-Wei Xuan. Clinical studies have shown that PSORI-CM02 is safe and effective for the treatment of psoriasis. However, its anti-psoriatic mechanisms remain to be further explored. In this study, we investigated the effects of PSORI-CM02 on angiogenesis in the skin and the underlying mechanisms in IL-17A-stimulated human umbilical vein endothelial cells (HUVECs) and a murine model of imiquimod (IMQ)-induced psoriasis. In vitro, PSORI-CM02 significantly inhibited the proliferation and migration of IL-17A-stimulated HUVECs in a dose-dependent manner. Further, it markedly regulated the antioxidative/oxidative status and inflammation; suppressed the expression of VEGF, VEGFR1, VEGFR2, ANG1, and HIF-1α; and reduced the phosphorylation of MAPK signalling pathway components in IL-17A-stimulated HUVECs. In vivo studies showed that PSORI-CM02 markedly reduced angiogenesis in the skin of mice with IMQ-induced psoriasis, while significantly rebalancing antioxidant/oxidant levels; inhibiting the production of IL-6, TNF-α, IL-17A, and IL-17F; and repressing the synthesis of angiogenic mediators. In addition, PSORI-CM02 markedly reduced the activation of the MAPK signalling pathway in psoriatic skin tissue. Taken together, our results demonstrated that PSORI-CM02 inhibited psoriatic angiogenesis by reducing the oxidative status and inflammation, suppressing the expression of angiogenesis-related molecules, and inhibiting the activation of the MAPK signalling pathway in vitro and in vivo.
Collapse
Affiliation(s)
- Yue Lu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Yang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junhong Zhang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyu Zhang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changju Ma
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojuan Tang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Li
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianan Wei
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiming Chen
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuanjian Lu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Han
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
13
|
Zhu WJ, Li P, Wang L, Xu YC. Hypoxia-inducible factor-1: A potential pharmacological target to manage psoriasis. Int Immunopharmacol 2020; 86:106689. [DOI: 10.1016/j.intimp.2020.106689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/27/2020] [Accepted: 06/06/2020] [Indexed: 12/16/2022]
|
14
|
Xue Y, Liu Y, Bian X, Zhang Y, Li Y, Zhang Q, Yin M. miR-205-5p inhibits psoriasis-associated proliferation and angiogenesis: Wnt/β-catenin and mitogen-activated protein kinase signaling pathway are involved. J Dermatol 2020; 47:882-892. [PMID: 32525225 DOI: 10.1111/1346-8138.15370] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/02/2020] [Indexed: 12/29/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease, and the mechanism remains unknown. The present study found that the level of miR-205-5p was downregulated in psoriatic skin tissues. miR-205-5p inhibited proliferation in HaCaT cells. miR-205-5p impaired proliferation, migration and tube formation in human umbilical vein endothelial cells. Angiopoietin (Ang)-2, vascular endothelial growth factor (VEGFA) and bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) were confirmed as the targets of miR-205-5p. Moreover, miR-205-5p suppressed the phosphorylation of p38 and extracellular regulated protein kinase, and inhibited expression level of β-catenin. In vivo, miR-205-5p significantly alleviated imiquimod (IMQ)-induced psoriasis in mice, and deactivated mitogen-activated protein kinase (MAPK) and Wnt/β-catenin pathways. In summary, we demonstrated that miR-205-5p alleviated IMQ-induced psoriasis in mice by restraining epidermal hyperproliferation and excessive neovascularization. miR-205-5p may play its roles by targeting Ang-2, VEGFA and BAMBI, and deactivating the Wnt/β-catenin and MAPK signaling pathways. These findings may provide a potential therapeutic target for clinical treatment of psoriasis.
Collapse
Affiliation(s)
- Yadong Xue
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Dermatology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingying Liu
- Department of Pathology, Heilongjiang Provincial Hospital, Harbin, China
| | - Xiaohui Bian
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South University, Changsha, China
| | - Yu Zhang
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuzhen Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qitao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingzhu Yin
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
15
|
Expression and Characterization of Human Vascular Endothelial Growth Factor Produced in SiHa Cells Transduced with Adenoviral Vector. Protein J 2019; 38:693-703. [DOI: 10.1007/s10930-019-09867-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
16
|
Min M, Yan BX, Wang P, Landeck L, Chen JQ, Li W, Cai SQ, Zheng M, Man XY. Rottlerin as a therapeutic approach in psoriasis: Evidence from in vitro and in vivo studies. PLoS One 2017; 12:e0190051. [PMID: 29272319 PMCID: PMC5741235 DOI: 10.1371/journal.pone.0190051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 12/07/2017] [Indexed: 01/09/2023] Open
Abstract
Rottlerin is a natural polyphenolic compound that was initially indicated as a PKCδ inhibitor. However, it was recently revealed that it may target a number of molecules and have biological effects on various cell types and is considered as a possible agent for tumor and cell proliferative diseases. Psoriasis is a chronic inflammatory cutaneous disorder with undefined etiology and is characterized by abnormal cellular proliferation, angiogenesis, and inflammation. Therefore, this paper investigates the regulatory effects of rottlerin on normal human epidermal keratinocytes (NHEKs) and imiquimod (IMQ)-induced psoriasiform (IPI) lesions. In vitro results showed that rottlerin inhibited cell proliferation in NHEKs through growth arrest and NFκB inhibition. It may also induce apoptosis in an autophagy-dependent pathway. We found that rottlerin inhibited human microvascular endothelial cells tube formation on matrigel. Rottlerin also decreased the cell senescence of keratinocytes and intracellular ROS generation, which indicated its antioxidant effect. We also showed that rottlerin affects the expression of keratinocyte proliferation biomarkers. In 12-O-tetradecanoylphorbol13-acetate (TPA)-induced keratinocytes, rottlerin significantly inhibited the expression of the induced pro-inflammatory cytokines in keratinocytes. An animal experiment provided the corresponding evidence based on this evidence in vitro, by using IPI model, we found that rottlerin could relieve the psoriasiform of BALB/c mice by inhibiting keratinocyte proliferation, inflammatory cell infiltration, and vascular proliferation. In conclusion, our results suggest that rottlerin may prove useful in the development of therapeutic agents against psoriasis. However, the deep mechanism still requires further study.
Collapse
Affiliation(s)
- Min Min
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xi Yan
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Wang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lilla Landeck
- Ernst von Bergmann General Hospital, Teaching Hospital of Charité– Humboldt University, Potsdam, Germany
| | - Jia-Qi Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- * E-mail: (MZ); (XYM)
| | - Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- * E-mail: (MZ); (XYM)
| |
Collapse
|
17
|
Hu SCS, Lan CCE. Psoriasis and Cardiovascular Comorbidities: Focusing on Severe Vascular Events, Cardiovascular Risk Factors and Implications for Treatment. Int J Mol Sci 2017; 18:ijms18102211. [PMID: 29065479 PMCID: PMC5666891 DOI: 10.3390/ijms18102211] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022] Open
Abstract
Psoriasis is a common and chronic inflammatory disease of the skin. It may impair the physical and psychosocial function of patients and lead to decreased quality of life. Traditionally, psoriasis has been regarded as a disease affecting only the skin and joints. More recently, studies have shown that psoriasis is a systemic inflammatory disorder which can be associated with various comorbidities. In particular, psoriasis is associated with an increased risk of developing severe vascular events such as myocardial infarction and stroke. In addition, the prevalence rates of cardiovascular risk factors are increased, including hypertension, diabetes mellitus, dyslipidemia, obesity, and metabolic syndrome. Consequently, mortality rates have been found to be increased and life expectancy decreased in patients with psoriasis, as compared to the general population. Various studies have also shown that systemic treatments for psoriasis, including methotrexate and tumor necrosis factor-α inhibitors, may significantly decrease cardiovascular risk. Mechanistically, the presence of common inflammatory pathways, secretion of adipokines, insulin resistance, angiogenesis, oxidative stress, microparticles, and hypercoagulability may explain the association between psoriasis and cardiometabolic disorders. In this article, we review the evidence regarding the association between psoriasis and cardiovascular comorbidities, focusing on severe vascular events, cardiovascular risk factors and implications for treatment.
Collapse
Affiliation(s)
- Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Cheng-Che E Lan
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| |
Collapse
|
18
|
Ibba-Manneschi L, Rosa I, Manetti M. Telocytes in Chronic Inflammatory and Fibrotic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 913:51-76. [PMID: 27796880 DOI: 10.1007/978-981-10-1061-3_4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Telocytes are a peculiar stromal (interstitial) cell type implicated in tissue homeostasis and development, as well as in the pathophysiology of several disorders. Severe damage and reduction of telocytes have been reported during fibrotic remodeling of multiple organs in various diseases, including scleroderma, Crohn's disease, ulcerative colitis, and liver fibrosis, as well as in chronic inflammatory lesions like those of primary Sjögren's syndrome and psoriasis. Owing to their close relationship with stem cells, telocytes are also supposed to contribute to tissue repair/regeneration. Indeed, telocytes are universally considered as "connecting cells" mostly oriented to intercellular signaling. On the basis of recent promising experimental findings, in the near future, telocyte transplantation might represent a novel therapeutic opportunity to control the evolution of chronic inflammatory and fibrotic diseases. Notably, there is evidence to support that telocytes could help in preventing abnormal activation of immune cells and fibroblasts, as well as in attenuating the altered matrix organization during the fibrotic process. By targeting telocytes alone or in tandem with stem cells, we might be able to promote regeneration and prevent the evolution to irreversible tissue injury. Besides exogenous transplantation, exploring pharmacological or non-pharmacological methods to enhance the growth and/or survival of telocytes could be an additional therapeutic strategy for many disorders.
Collapse
Affiliation(s)
- Lidia Ibba-Manneschi
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy.
| |
Collapse
|
19
|
Niu X, Chang W, Liu R, Hou R, Li J, Wang C, Li X, Zhang K. Expression of pro-angiogenic genes in mesenchymal stem cells derived from dermis of patients with psoriasis. Int J Dermatol 2016; 55:e280-8. [PMID: 26748901 DOI: 10.1111/ijd.13197] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 07/02/2015] [Accepted: 08/24/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Recent experimental studies revealed that angiogenesis and lymphangiogenesis are closely related to psoriasis. Our microarray analysis suggested that the pro-angiogenic genes platelet endothelial cell adhesion molecule-1 (PECAM1), facio-genital dysplasia-5 (FGD5), prostaglandin-endoperoxide synthase-1 (PTGS1), melanoma cell adhesion molecule (MCAM), vasohibin-2 (VASH2), and stabilin-1 (STAB1) are differentially expressed in dermal mesenchymal stem cells in psoriasis. OBJECTIVES The aim of this study was to investigate the mRNA and protein expression of PECAM1, FGD5, PTGS1, MCAM, VASH2, and STAB1 for angiogenesis and the possible mechanisms in psoriasis. METHODS We studied 12 patients with plaque psoriasis and 14 healthy controls matched for age and sex. Dermal mesenchymal stem cells were expanded, passaged, and identified by cellular morphology, immunophenotyping, and multipotential differentiation. The mRNA and protein expression of the above-mentioned six genes were confirmed by quantitative real-time reverse transcription-polymerase chain reaction and Western blotting. RESULTS The significantly decreased expression of PECAM1, PTGS1, FGD5, and MCAM at both mRNA and protein level (except VASH2 and STAB1) were demonstrated in mesenchymal stem cells from psoriatic skin lesions compared with non-lesional from healthy controls. CONCLUSIONS We provide the first report that pro-angiogenic genes PECAM1, PTGS1, FGD5, and MCAM rather than VASH2 and STAB1 may be play a vital role in pathological dermal angiogenesis disorders of psoriasis. Therefore, anti-angiogenesis is attractive and offers future potential for application in patients with psoriasis.
Collapse
Affiliation(s)
- Xuping Niu
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - WenJuan Chang
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Ruifeng Liu
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Ruixia Hou
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Junqin Li
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Chunfang Wang
- Laboratory Animal Center, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xinhua Li
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Kaiming Zhang
- Institute of Dermatology, Taiyuan City Central Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
20
|
Niu X, Chang W, Liu R, Hou R, Li J, Wang C, Li X, Zhang K. mRNA and protein expression of the angiogenesis-related genes EDIL3, AMOT and ECM1 in mesenchymal stem cells in psoriatic dermis. Clin Exp Dermatol 2015; 41:533-40. [PMID: 26644074 DOI: 10.1111/ced.12783] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Dermal microvasculature expansion and angiogenesis are prominent in psoriasis. Our previous microarray study showed that the angiogenesis-related genes EDIL3 (epidermal growth factor-like repeats and discoidin I-like domains 3), AMOT (angiomotin) and ECM1 (extracellular matrix protein 1), had high expression levels in dermal mesenchymal stem cells (DMSCs) from psoriatic skin lesions. AIM To investigate the mRNA and protein expressions of EDIL3, AMOT and ECM1 in DMSCs derived from psoriatic skin in order to better determine the molecular mechanisms of angiogenesis in the skin. METHODS DMSCs from 12 patients with psoriasis and 14 healthy controls (HCs) were cultured to passage 3, and identified by morphology, immunophenotype and multipotential differentiation. The mRNA and protein expressions of EDIL3, AMOT, and ECM1 in the DMSCs were determined using real-time reverse transcription PCR and western blotting. RESULTS DMSCs displayed spindle-like morphology and surface protein expression, and were able to differentiate into osteoblasts, chondrocytes and adipocytes. mRNA expression analysis showed that EDIL3, AMOT and ECM1 were expressed at 2.54-fold, 2.11-fold, and 1.90-fold higher levels, respectively, in psoriatic DMSCs compared with HC DMSCs (all P < 0.05). Protein analysis showed significantly (all P < 0.01) higher concentrations of EDIL3, AMOT and ECM1in the psoriasis group than in the HC group. CONCLUSIONS Our data demonstrate for the first time that expression of EDIL3, AMOT and ECM1 is altered in DMSCs in psoriasis, suggesting that EDIL3, AMOT and ECM1 are involved in the excessive angiogenesis and vasodilation observed in psoriasis.
Collapse
Affiliation(s)
- X Niu
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - W Chang
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - R Liu
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - R Hou
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - J Li
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - C Wang
- Laboratory Animal Center, Shanxi Medical University, Taiyuan, Shanxi, China
| | - X Li
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| | - K Zhang
- Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
21
|
Elshabrawy HA, Chen Z, Volin MV, Ravella S, Virupannavar S, Shahrara S. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis 2015; 18:433-48. [PMID: 26198292 PMCID: PMC4879881 DOI: 10.1007/s10456-015-9477-2] [Citation(s) in RCA: 362] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/14/2015] [Indexed: 12/31/2022]
Abstract
Angiogenesis is the formation of new capillaries from pre-existing vasculature, which plays a critical role in the pathogenesis of several inflammatory autoimmune diseases such as rheumatoid arthritis (RA), spondyloarthropathies, psoriasis, systemic lupus erythematosus, systemic sclerosis, and atherosclerosis. In RA, excessive migration of circulating leukocytes into the inflamed joint necessitates formation of new blood vessels to provide nutrients and oxygen to the hypertrophic joint. The dominance of the pro-angiogenic factors over the endogenous angiostatic mediators triggers angiogenesis. In this review article, we highlight the underlying mechanisms by which cells present in the RA synovial tissue are modulated to secrete pro-angiogenic factors. We focus on the significance of pro-angiogenic factors such as growth factors, hypoxia-inducible factors, cytokines, chemokines, matrix metalloproteinases, and adhesion molecules on RA pathogenesis. As pro-angiogenic factors are primarily produced from RA synovial tissue macrophages and fibroblasts, we emphasize the key role of RA synovial tissue lining layer in maintaining synovitis through neovascularization. Lastly, we summarize the specific approaches utilized to target angiogenesis. We conclude that the formation of new blood vessels plays an indispensable role in RA progression. However, since the function of several pro-angiogenic mediators is cross regulated, discovering novel approaches to target multiple cascades or selecting an upstream cascade that impairs the activity of a number of pro-angiogenic factors may provide a promising strategy for RA therapy.
Collapse
Affiliation(s)
- Hatem A Elshabrawy
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Zhenlong Chen
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, 60515, USA
| | - Shalini Ravella
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Shanti Virupannavar
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA.
| |
Collapse
|
22
|
Liu Y, Yang G, Zhang J, Xing K, Dai L, Cheng L, Liu J, Deng J, Shi G, Li C, Su X, Zhang S, Yang Y, Li J, Yu D, Xiang R, Wei Y, Deng H. Anti-TNF-α monoclonal antibody reverses psoriasis through dual inhibition of inflammation and angiogenesis. Int Immunopharmacol 2015; 28:731-43. [PMID: 26263167 DOI: 10.1016/j.intimp.2015.07.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/28/2015] [Accepted: 07/27/2015] [Indexed: 02/05/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) antagonists have shown remarkable efficacy in psoriasis; however, the precise mechanisms of action of TNF-α blocking agents mainly focus on their neutralizing TNF-α and its anti-inflammatory effects. In this study, we generated a humanized anti-TNF-α monoclonal antibody (IBI303) and suggested a potential mechanism of anti-TNF-α therapy for psoriasis. The results of SPR and ELISA indicated that IBI303 has a good affinity to TNF-α. In vitro, it could suppress TNF-α-induced cytotoxicity in WEHI164 cells. In vivo, administration of IBI303 to K14-VEGF transgenic mice led to a significant treatment efficiency in psoriasis in a dose-dependent manner. IHC staining and cytokines-ELISA indicated that TNF-α inhibition strongly reduced inflammatory cells infiltration and pro-inflammatory cytokines release, accompanied by suppression of inflamed dermal blood vessels. Mechanistically, in order to explain the anti-angiogenesis effect of anti-TNF-α antibody, the production of cytokine in macrophage conditional medium was measured by ELISA. The result indicated that the massive secretion of TNF-α stimulated by LPS in RAW264.7 cell supernatant was markedly neutralized in a dose-response manner by IBI303, moreover, the expression of NF-κB p65 was down-regulated. Mouse endothelial cell tube formation assay showed that anti-TNF-α could inhibit blood vessels formation directly and indirectly. Collectively, our study suggested a kind of antipsoriatic mechanism of TNF-α inhibitors that is the dual inhibition of inflammation and angiogenesis.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Guoyou Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Junfeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Kaiyan Xing
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Junli Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Jie Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Chunlei Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China; Chongqing Tree Gorges Medical College, Faculty of Basic Medicine, Department of Biochemistry, People's Republic of China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Shuang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Jia Li
- Innovent Biologics, Inc., Suzhou, Jiangsu, People's Republic of China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China; Innovent Biologics, Inc., Suzhou, Jiangsu, People's Republic of China
| | - Rong Xiang
- Department of Immunology, Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
23
|
Li J, Li X, Hou R, Liu R, Zhao X, Dong F, Wang C, Yin G, Zhang K. Psoriatic T cells reduce epidermal turnover time and affect cell proliferation contributed from differential gene expression. J Dermatol 2015; 42:874-80. [PMID: 26046687 DOI: 10.1111/1346-8138.12961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Junqin Li
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Xinhua Li
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Ruixia Hou
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Ruifeng Liu
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Xincheng Zhao
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Feng Dong
- Department of Dermatology; Changzhi City Second People's Hospital; Changzhi China
| | - Chunfang Wang
- Laboratory Animal Center; Shanxi Medical University; Taiyuan China
| | - Guohua Yin
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| | - Kaiming Zhang
- Institute of Dermatology; Taiyuan City Center Hospital; Taiyuan China
| |
Collapse
|
24
|
Manole CG, Gherghiceanu M, Simionescu O. Telocyte dynamics in psoriasis. J Cell Mol Med 2015; 19:1504-19. [PMID: 25991475 PMCID: PMC4511349 DOI: 10.1111/jcmm.12601] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/03/2015] [Indexed: 12/18/2022] Open
Abstract
The presence of telocytes (TCs) as distinct interstitial cells was previously documented in human dermis. TCs are interstitial cells completely different than dermal fibroblasts. TCs are interconnected in normal dermis in a 3D network and may be involved in skin homeostasis, remodelling, regeneration and repair. The number, distribution and ultrastructure of TCs were recently shown to be affected in systemic scleroderma. Psoriasis is a common inflammatory skin condition (estimated to affect about 0.1-11.8% of population), a keratinization disorder on a genetic background. In psoriasis, the dermis contribution to pathogenesis is frequently eclipsed by remarkable epidermal phenomena. Because of the particular distribution of TCs around blood vessels, we have investigated TCs in the dermis of patients with psoriasis vulgaris using immunohistochemistry (IHC), immunofluorescence (IF), and transmission electron microscopy (TEM). IHC and IF revealed that CD34/PDGFRα-positive TCs are present in human papillary dermis. More TCs were present in the dermis of uninvolved skin and treated skin than in psoriatic dermis. In uninvolved skin, TEM revealed TCs with typical ultrastructural features being involved in a 3D interstitial network in close vicinity to blood vessels in contact with immunoreactive cells in normal and treated skin. In contrast, the number of TCs was significantly decreased in psoriatic plaque. The remaining TCs demonstrated multiple degenerative features: apoptosis, membrane disintegration, cytoplasm fragmentation and nuclear extrusion. We also found changes in the phenotype of vascular smooth muscle cells in small blood vessels that lost the protective envelope formed by TCs. Therefore, impaired TCs could be a 'missed' trigger for the characteristic vascular pathology in psoriasis. Our data explain the mechanism of Auspitz's sign, the most pathognomonic clinical sign of psoriasis vulgaris. This study offers new insights on the cellularity of psoriatic lesions and we suggest that TCs should be considered new cellular targets in forthcoming therapies.
Collapse
Affiliation(s)
- C G Manole
- Department of Cell Biology and Histology, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.,Laboratory of Ultrastructural Research, 'Victor Babeş' National Institute of Pathology, Bucharest, Romania
| | - Mihaela Gherghiceanu
- Laboratory of Ultrastructural Research, 'Victor Babeş' National Institute of Pathology, Bucharest, Romania
| | - Olga Simionescu
- Department of Dermatology, Colentina University Hospital, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
25
|
Pal HC, Chamcheu JC, Adhami VM, Wood GS, Elmets CA, Mukhtar H, Afaq F. Topical application of delphinidin reduces psoriasiform lesions in the flaky skin mouse model by inducing epidermal differentiation and inhibiting inflammation. Br J Dermatol 2014; 172:354-64. [PMID: 25533330 DOI: 10.1111/bjd.13513] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease characterized by hyperproliferation and aberrant keratinocyte differentiation. We have shown that treatment of reconstituted human skin with delphinidin, an anthocyanidin, present in pigmented fruits and vegetables, increased the expression and processing of caspase-14, which is involved in cornification. Delphinidin also increases the expression of epidermal differentiation marker proteins. OBJECTIVES To determine whether topical application of delphinidin can modulate pathological markers of psoriasiform lesions in flaky skin mice and if this is associated with increased epidermal differentiation and a reduction in proliferation and inflammation. METHODS Five-week-old female homozygous flaky skin mice (fsn/fsn) were treated topically with delphinidin (0·5 mg cm(-2) and 1 mg cm(-2) skin areas, respectively), five times a week, up to 14 weeks of age. RESULTS Treatment of flaky skin mice with delphinidin resulted in a reduction in (i) pathological markers of psoriasiform lesions; (ii) infiltration of inflammatory cells; and (iii) mRNA and protein expression of inflammatory cytokines. Delphinidin treatment also increased the expression and processing of caspase-14, and expression of filaggrin, loricrin, keratin-1 and keratin-10. Furthermore, there was a decrease in the expression of markers for cell proliferation (proliferating cell nuclear antigen and keratin-14) and modulation of tight junction proteins (occludin and claudin-1). In addition, delphinidin treatment increased the expression of activator protein-1 transcription factor proteins (JunB, JunD, Fra1 and Fra2). CONCLUSIONS Delphinidin could be a promising agent for treatment of psoriasis and other hyperproliferative skin disorders.
Collapse
Affiliation(s)
- H C Pal
- Department of Dermatology, University of Alabama at Birmingham, 1670 University Blvd, Birmingham, 35294, AL, U.S.A
| | | | | | | | | | | | | |
Collapse
|
26
|
Ayata RE, Bouhout S, Auger M, Pouliot R. Study of in vitro capillary-like structures in psoriatic skin substitutes. Biores Open Access 2014; 3:197-205. [PMID: 25371856 PMCID: PMC4215329 DOI: 10.1089/biores.2014.0022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis is one of the important hallmarks of psoriasis. The extension of the superficial microvascular structure and activated pro-angiogenic mediators in psoriasis seem to be important factors involved in the pathology. According to the changes of superficial microvasculature in psoriatic lesions, anti-angiogenic treatment could be a promising therapeutic strategy for psoriasis. The aim of this study was to construct an in vitro vascularized psoriatic skin substitute for fundamental research. Psoriatic fibroblasts and keratinocytes were isolated from psoriatic plaque biopsies, while healthy fibroblasts and keratinocytes, as well as microvascular endothelial cells, were isolated from healthy skin biopsies of cosmetic breast surgery. Psoriatic and healthy skin substitutes with and without endothelial cells were produced using the self-assembly approach. Afterward the substitutes were examined by histology, immunofluorescence studies, and three-dimensional (3D) confocal microscopy. Histological analysis and immunofluorescence staining of specific markers for endothelial cells (von Willebrand, PECAM-1 [CD31], and VE-cadherin [CD144]) and basement membrane component (collagen IV) demonstrated that endothelial cells have the ability to form capillary-like tubes. Moreover, the 3D branched structure of the capillary-like structures and an eagle eye view of them were observed by confocal microscopy. Also the semiquantification of capillary-like tubes (CLTs) was carried out with a 3D eagle eye view of substitutes, and more CLTs were observed in psoriatic substitutes. These results suggest that it is possible to observe 3D capillary-like structures in the self-assembled psoriatic skin substitutes, which could become a good in vitro testing model for anti-angiogenic drug research, and facilitate the study of this complex pathology, which links angiogenesis to its development.
Collapse
Affiliation(s)
- Raif Eren Ayata
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval , Québec, Canada . ; Division of Regenerative Medicine, CHU de Québec Research Centre , Québec, Canada . ; Faculté de Pharmacie, Université Laval , Québec, Canada
| | - Sara Bouhout
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval , Québec, Canada . ; Division of Regenerative Medicine, CHU de Québec Research Centre , Québec, Canada
| | - Michèle Auger
- Département de Chimie, PROTEO, CERMA, Université Laval , Québec, Canada
| | - Roxane Pouliot
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval , Québec, Canada . ; Division of Regenerative Medicine, CHU de Québec Research Centre , Québec, Canada . ; Faculté de Pharmacie, Université Laval , Québec, Canada
| |
Collapse
|
27
|
Zeng L, Cui J, Wu H, Lu Q. The emerging role of circulating microRNAs as biomarkers in autoimmune diseases. Autoimmunity 2014; 47:419-29. [PMID: 24953646 DOI: 10.3109/08916934.2014.929667] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The highly conserved RNAs known as microRNAs (miRNAs) are a class of small, single-stranded, non-coding RNAs that play a critical role in the regulation of host genome expression at the posttranscriptional level. MiRNA-mediated gene regulation is vital for normal cellular functions, such as the cell differentiation, proliferation and apoptosis, and nearly one-third of human messenger RNAs might be miRNA targets. Increasing evidence has suggested that miRNAs play a critical role in the regulating the immune system and preventing autoimmune disorders. Circulating miRNAs, which can be easily detected by a non-invasive methods, have been proven to be able to distinguish diseased individuals from healthy subjects. In addition, these circulating miRNAs have relatively high sensitivity and specificity and thus have been developed as biomarkers for the diagnosis and monitoring of human diseases. To date, nearly 100 circulating miRNAs have been proven to be biomarkers for various diseases, and this number continues to rise. This review aims to summarize the most promising identified circulating miRNAs as potential biomarkers in autoimmune diseases and to discuss current challenges and future directions in the field.
Collapse
Affiliation(s)
- Li Zeng
- Department of Dermatology, The 2nd Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenetics , Changsha , China
| | | | | | | |
Collapse
|
28
|
Forkel S, Schön M, Hildmann A, Claßen A, John SM, Danker K, Schön MP. Inositoylated platelet-activating factor (Ino-C2-PAF) modulates dynamic lymphocyte-endothelial cell interactions and alleviates psoriasis-like skin inflammation in two complementary mouse models. J Invest Dermatol 2014; 134:2510-2520. [PMID: 24714204 DOI: 10.1038/jid.2014.170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/23/2014] [Accepted: 03/14/2014] [Indexed: 02/08/2023]
Abstract
Psoriasis, a tumor necrosis factor alpha (TNFα)-governed inflammatory disorder with prominent dysregulation of cutaneous vascular functions, has evolved into a model disorder for studying anti-inflammatory therapies. We present experimental in vitro and in vivo data on 1-O-octadecyl-2-O-(2-(myo-inositolyl)-ethyl)-sn-glycero-3-(R/S)-phosphatidyl-choline (Ino-C2-PAF), the lead compound of a class of synthetic glycosylated phospholipids, in anti-inflammatory therapy. Ino-C2-PAF strongly induced apoptosis only in TNFα-stimulated, but not in untreated human vascular endothelial cells. Moreover, TNFα-induced endothelial adhesion molecules that mediated the rolling and firm adhesion of leukocytes (vascular cell adhesion protein-1 (VCAM-1), E-selectin, and ICAM-1) were selectively downregulated by Ino-C2-PAF. Similarly, expression of L-selectin, VCAM-1 receptor α4β1 integrin , and lymphocyte function-associated antigen-1 on human peripheral blood mononuclear cells was reduced without induction of apoptosis. Functionally, these changes were accompanied by significant impairment of rolling and adhesion of human peripheral blood lymphocytes on TNFα-activated endothelial cells in a dynamic flow chamber system. When the therapeutic potential of Ino-C2-PAF was assessed in two complementary mouse models of psoriasis, K5.hTGFβ1 transgenic and JunB/c-Jun-deficient mice, Ino-C2-PAF led to significant alleviation of the clinical symptoms and normalized the pathological cutaneous changes including vascularization. There were no overt adverse effects. These findings suggested that Ino-C2-PAF is a potential candidate in the therapy of inflammatory skin diseases that include abnormal vascular functions.
Collapse
Affiliation(s)
- Susann Forkel
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | - Margarete Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany
| | - Annette Hildmann
- Institute of Biochemistry, Charité University Medical Center, Berlin, Germany
| | - Anna Claßen
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany
| | - Swen-Malte John
- Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany; Department of Dermatology, Environmental Medicine and Health Care Theory, University of Osnabrück, Osnabrück, Germany
| | - Kerstin Danker
- Institute of Biochemistry, Charité University Medical Center, Berlin, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg August University, Göttingen, Germany; Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Osnabrück, Germany.
| |
Collapse
|
29
|
Meki ARMA, Al-Shobaili H. Serum vascular endothelial growth factor, transforming growth factor β1, and nitric oxide levels in patients with psoriasis vulgaris: their correlation to disease severity. J Clin Lab Anal 2014; 28:496-501. [PMID: 24659464 DOI: 10.1002/jcla.21717] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 10/28/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF), transforming growth factor β1 (TGF-β1), and nitric oxide (NO) have been reported to be contributory factors to the pathogenesis of psoriasis vulgaris. In the current study, we aimed to investigate the association between the levels of VEGF, TGF-β1, and NO and psoriasis severity (as expressed by psoriasis area severity index, PASI). METHODS Fifty-eight patients with psoriasis vulgaris and twenty-two controls were included in the study. The serum levels of VEGF and TGF-β1 were estimated by ELISA technique. The serum levels of NO were determined by colorimetric method. RESULTS The serum levels of VEGF, TGF-β1, and NO were significantly higher in patients than controls. Moreover, the serum levels of the studied biochemical variables in patients with severe disease activity were significantly higher than mild cases. The duration of disease showed significant positive correlations with each VEGF (r = 0.35, P < 0.01) and TGF-β1 (r = 0.41, P < 0.05). In addition, the PASI score was significantly positively correlated with VEGF (r = 0.65, P < 0.001), TGF-β1 (r = 0.31, P < 0.05), and NO (r = 0.51, P < 0.001). CONCLUSION These findings suggest an association between psoriasis disease severity and serum levels of VEGF, TGF-β1, and NO, which can be recognized as markers of the psoriasis severity. The modulation of their production may represent a therapeutic potential strategy for psoriasis.
Collapse
Affiliation(s)
- Abdel-Raheim M A Meki
- Department of Medical Biochemistry, College of Medicine, Qassim University, Almlaida, Kingdom of Saudi Arabia
| | | |
Collapse
|
30
|
Willerslev-Olsen A, Litvinov IV, Fredholm SM, Petersen DL, Sibbesen NA, Gniadecki R, Zhang Q, Bonefeld CM, Wasik MA, Geisler C, Zhou Y, Woetmann A, Sasseville D, Krejsgaard T, Odum N. IL-15 and IL-17F are differentially regulated and expressed in mycosis fungoides (MF). Cell Cycle 2014; 13:1306-12. [PMID: 24621498 DOI: 10.4161/cc.28256] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Skin lesions from mycosis fungoides (MF) patients display an increased expression of interleukin-15 (IL-15), IL-17F, and other cytokines implicated in inflammation and malignant cell proliferation in cutaneous T-cell lymphoma (CTCL). In the leukemic variant of CTCL, Sézary syndrome (SS), IL-2 and IL-15 trigger activation of the Jak-3/STAT3 pathway and transcription of IL17A gene, whereas it is unknown what causes IL-15 expression, Jak3/STAT3 activation, and production of IL-17F in MF. Here, we studied the expression and regulation of IL-15 and its relation to IL-17F in MF cell lines and skin lesions from 60 MF patients. We show that: (1) the spontaneous IL-15 mRNA expression is resistant to Jak3 and STAT3 inhibitors at concentrations that profoundly inhibit STAT3 activation and IL-17F mRNA expression; (2) anti-IL-15 antibody blocks STAT3 activation induced by exogenous IL-15 in non-malignant MF T cells, whereas the spontaneous STAT3 activation and IL-17F expression in malignant T cells is not inhibited; (3) patients display heterogeneous IL-15/IL-17F mRNA expression patterns in skin lesions; and (4) IL-15 expression (in contrast to IL-17F) is not associated with progressive disease. Taken together, these findings indicate that IL-15 and IL-17F are differentially regulated and expressed in MF. We propose that IL-15 and IL-17F are markers for different inflammatory environments and play distinct roles in the development and progression of MF.
Collapse
Affiliation(s)
- Andreas Willerslev-Olsen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Ivan V Litvinov
- Division of Dermatology; McGill University Health Centre; Montréal, Quebec, Canada
| | - Simon M Fredholm
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - David L Petersen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Nina A Sibbesen
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Robert Gniadecki
- Departmen of Dermatology; Copenhagen University Hospital; Bispebjerg, Copenhagen, Denmark
| | - Qian Zhang
- Department of Pathology and Laboratory Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Charlotte M Bonefeld
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Carsten Geisler
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Youwen Zhou
- Department of Dermatology and Skin Science; University of British Columbia; Vancouver, British Columbia, Canada
| | - Anders Woetmann
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Denis Sasseville
- Division of Dermatology; McGill University Health Centre; Montréal, Quebec, Canada
| | - Thorbjørn Krejsgaard
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| | - Niels Odum
- Department of International Health, Immunology, and Microbiology; University of Copenhagen; Copenhagen, Denmark
| |
Collapse
|
31
|
Affiliation(s)
- Houda Haouas
- Department of Biological and Chemical EngineeringNational Institute of Applied Sciences and Technology, Tunis, Tunisia
| |
Collapse
|
32
|
Villalta SA, Lang J, Kubeck S, Kabre B, Szot GL, Calderon B, Wasserfall C, Atkinson MA, Brekken RA, Pullen N, Arch RH, Bluestone JA. Inhibition of VEGFR-2 reverses type 1 diabetes in NOD mice by abrogating insulitis and restoring islet function. Diabetes 2013; 62:2870-8. [PMID: 23835340 PMCID: PMC3717875 DOI: 10.2337/db12-1619] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The dysregulation of receptor tyrosine kinases (RTKs) in multiple cell types during chronic inflammation is indicative of their pathogenic role in autoimmune diseases. Among the many RTKs, vascular endothelial growth factor receptor (VEGFR) stands out for its multiple effects on immunity, vascularization, and cell migration. Herein, we examined whether VEGFR participated in the pathogenesis of type 1 diabetes (T1D) in nonobese diabetic (NOD) mice. We found that RTK inhibitors (RTKIs) and VEGF or VEGFR-2 antibodies reversed diabetes when administered at the onset of hyperglycemia. Increased VEGF expression promoted islet vascular remodeling in NOD mice, and inhibition of VEGFR activity with RTKIs abrogated the increase in islet vascularity, impairing T-cell migration into the islet and improving glucose control. Metabolic studies confirmed that RTKIs worked by preserving islet function, as treated mice had improved glucose tolerance without affecting insulin sensitivity. Finally, examination of human pancreata from patients with T1D revealed that VEGFR-2 was confined to the islet vascularity, which was increased in inflamed islets. Collectively, this work reveals a previously unappreciated role for VEGFR-2 signaling in the pathogenesis of T1D by controlling T-cell accessibility to the pancreatic islets and highlights a novel application of VEGFR-2 antagonists for the therapeutic treatment of T1D.
Collapse
Affiliation(s)
- S. Armando Villalta
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Jiena Lang
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Samantha Kubeck
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Beniwende Kabre
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Gregory L. Szot
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Boris Calderon
- Division of Laboratory and Genomic Medicine; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Clive Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Rolf A. Brekken
- Division of Surgical Oncology, Department of Surgery and Department of Pharmacology, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nick Pullen
- Pfizer Global Research and Development, Cambridge, Massachusetts
| | - Robert H. Arch
- Pfizer Global Research and Development, Chesterfield, Missouri
| | - Jeffrey A. Bluestone
- Diabetes Center and the Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
33
|
Angiogenic and inflammatory properties of psoriatic arthritis. ISRN DERMATOLOGY 2013; 2013:630620. [PMID: 23819059 PMCID: PMC3683428 DOI: 10.1155/2013/630620] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 05/15/2013] [Indexed: 12/29/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy associated with psoriasis and included in seronegative spondyloarthropathy. PsA has several unique characteristics different from rheumatoid arthritis (RA), such as enthesopathy, dactylitis, and abnormal bone remodeling. As compared with synovitis of RA (pannus), proliferation of PsA synovium is mild and characterized by hypervascularity and increased infiltration of polymorphonuclear leukocytes in the synovial tissues. Angiogenesis plays a crucial role in cutaneous psoriasis, and several angiogenic factors such as vascular endothelial growth factor, interleukin-8, angiopoietin, tumor necrosis factor- α and transforming growth factor-β, are suggested to play an important role also in the pathophysiology of PsA. Further, IL-17 has various functions such as upregulation of proinflammatory cytokines, attraction of neutrophils, stimulation of keratinocytes, endothelial cell migration, and osteoclast formation via RANKL from activated synovial fibroblasts. Thus, IL-17 may be important in angiogenesis, fibrogenesis, and osteoclastogenesis in PsA. In this paper, roles of angiogenesis in the psoriatic synovium are discussed, which may strengthen the understanding of the pathogenesis of PsA.
Collapse
|
34
|
High serum levels of HIF-1α in psoriatic patients correlate with an over-expression of IL-6. Cytokine 2013; 62:38-9. [DOI: 10.1016/j.cyto.2013.02.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 02/05/2013] [Accepted: 02/23/2013] [Indexed: 10/27/2022]
|
35
|
Involvement of IL-9 in Th17-associated inflammation and angiogenesis of psoriasis. PLoS One 2013; 8:e51752. [PMID: 23335955 PMCID: PMC3546056 DOI: 10.1371/journal.pone.0051752] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 11/05/2012] [Indexed: 11/19/2022] Open
Abstract
It is thought that a Th1/Th17-weighted immune response plays a predominant role in the pathogenesis of psoriasis. Our findings now indicate a link between IL-9, a Th2 and Th9 cytokine, and Th17 pathway in psoriasis. In K5.hTGF-β1 transgenic mice, exhibiting a psoriasis-like phenotype, we found increased IL-9R and IL-9 expression in the skin and intradermal IL-9 injection induced Th17-related inflammation. IL-9 also promoted angiogenesis and VEGF and CD31 overexpression in mice in vivo and increased tube formation of human endothelial cells in vitro. Injecting anti-IL-9 antibody into K5.hTGF-β1 transgenic mice not only diminished inflammation (including skin infiltration by T cells, monocytes/macrophages, and mast cells) and angiogenesis but also delayed the psoriasis-like skin phenotype. Notably, injection of anti-psoriatic acting anti-IL-17 antibody reduced skin IL-9 mRNA and serum IL-9 protein levels in K5.hTGF-β1 transgenic mice and prevented IL-9-induced epidermal hyperplasia and inflammation of the skin of wild type mice. In addition, we observed that IL-9R expression in lesional skin from psoriasis patients was markedly higher than in healthy skin from control subjects. Moreover, IL-9 significantly enhanced IL-17A production by cultured human peripheral blood mononuclear cells or CD4+ T cells, especially in psoriasis patients. Thus, IL-9 may play a role in the development of psoriatic lesions through Th17-associated inflammation and angiogenesis.
Collapse
|
36
|
Guma M, Firestein GS. c-Jun N-Terminal Kinase in Inflammation and Rheumatic Diseases. Open Rheumatol J 2012; 6:220-31. [PMID: 23028407 PMCID: PMC3460413 DOI: 10.2174/1874312901206010220] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 01/03/2011] [Accepted: 07/13/2011] [Indexed: 01/24/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are members of the mitogen-activated protein kinase (MAPK) family and are activated by environmental stress. JNK is also activated by proinflammatory cytokines, such as TNF and IL-1, and Toll-like receptor ligands. This pathway, therefore, can act as a critical convergence point in immune system signaling for both adaptive and innate responses. Like other MAPKs, the JNKs are activated via the sequential activation of protein kinases that includes two dual-specificity MAP kinase kinases (MKK4 and MKK7) and multiple MAP kinase kinase kinases. MAPKs, including JNKs, can be deactivated by a specialized group of phosphatases, called MAP kinase phosphatases. JNK phosphorylates and regulates the activity of transcription factors other than c-Jun, including ATF2, Elk-1, p53 and c-Myc and non-transcription factors, such as members of the Bcl-2 family. The pathway plays a critical role in cell proliferation, apoptosis, angiogenesis and migration. In this review, an overview of the functions that are related to rheumatic diseases is presented. In addition, some diseases in which JNK participates will be highlighted.
Collapse
Affiliation(s)
- Monica Guma
- Division of Rheumatology, Allergy and Immunology, UC San Diego School of Medicine, La Jolla, CA, USA
| | | |
Collapse
|
37
|
Kwon MJ, Kim B, Lee YS, Kim TY. Role of superoxide dismutase 3 in skin inflammation. J Dermatol Sci 2012; 67:81-7. [DOI: 10.1016/j.jdermsci.2012.06.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/01/2022]
|
38
|
Abdelsaid MA, El-Remessy AB. S-glutathionylation of LMW-PTP regulates VEGF-mediated FAK activation and endothelial cell migration. J Cell Sci 2012; 125:4751-60. [PMID: 22854047 DOI: 10.1242/jcs.103481] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although promising, the ability to regulate angiogenesis through delivery of VEGF remains an unrealized goal. We have shown previously that physiological levels of peroxynitrite (1 µM) are required for a VEGF-mediated angiogenic response, yet the redox-regulated mechanisms that govern the VEGF signal remain unexplored. We assessed the impact of VEGF and peroxynitrite on modifying redox-state, the level of reduced-glutathione (GSH) and S-glutathionylation on regulation of the low molecular weight protein tyrosine phosphatase (LMW-PTP) and focal adhesion kinase (FAK), which are key mediators of VEGF-mediated cell migration. Stimulation of human microvascular endothelial (HME) cells with VEGF (20 ng/ml) or peroxynitrite (1 µM) caused an immediate and reversible negative-shift in the cellular redox-state and thiol oxidation of LMW-PTP, which culminated in cell migration. VEGF causes reversible S-glutathionylation of LMW-PTP, which inhibits its phosphorylation and activity, and causes the transient activation of FAK. Modulating the redox-state using decomposing peroxynitrite (FeTPPS, 2.5 µM) or the GSH-precursor [N-acetylcysteine (NAC), 1 mM] caused a positive-shift of the redox-state and prevented VEGF-mediated S-glutathionylation and oxidative inhibition of LMW-PTP. NAC and FeTPPS prevented the activation of FAK, its association with LMW-PTP and cell migration. Inhibiting LMW-PTP expression markedly enhanced FAK activation and cell migration. Although mild oxidative stress achieved by combining VEGF with 0.1-0.2 mM peroxynitrite augmented cell migration, an acute shift to oxidative stress achieved by combining VEGF with 0.5 mM peroxynitrite induced and sustained FAK activation, and LMW-PTP S-glutathionylation, resulting in LMW-PTP inactivation and inhibited cell migration. In conclusion, our findings demonstrate that a balanced redox-state is required for VEGF to facilitate reversible S-glutathionylation of LMW-PTP, FAK activation and endothelial cell migration. Shifting the redox-state to reductive stress or oxidative stress inhibited the VEGF-mediated angiogenic response.
Collapse
|
39
|
Delfino G, Di Costanzo L, Paulis A, Fabbrocini G, Monfrecola G. In vitro ultraviolet A irradiation decreases both release ability and gene-expression of vascular endothelial growth factor-A from mast cells. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2012; 28:165-8. [DOI: 10.1111/j.1600-0781.2012.00652.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Gabriele Delfino
- Division of Allergy and Clinical Immunology; University of Naples Federico II; Italy
| | | | - Amato Paulis
- Division of Allergy and Clinical Immunology; University of Naples Federico II; Italy
| | | | | |
Collapse
|
40
|
Calderari S, Chougnet C, Clemessy M, Kempf H, Corvol P, Larger E. Angiopoietin 2 alters pancreatic vascularization in diabetic conditions. PLoS One 2012; 7:e29438. [PMID: 22272235 PMCID: PMC3260141 DOI: 10.1371/journal.pone.0029438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/28/2011] [Indexed: 01/25/2023] Open
Abstract
Aims/hypothesis Islet vascularization, by controlling beta-cell mass expansion in response to increased insulin demand, is implicated in the progression to glucose intolerance and type 2 diabetes. We investigated how hyperglycaemia impairs expansion and differentiation of the growing pancreas. We have grafted xenogenic (avian) embryonic pancreas in severe combined immuno-deficient (SCID) mouse and analyzed endocrine and endothelial development in hyperglycaemic compared to normoglycaemic conditions. Methods 14 dpi chicken pancreases were grafted under the kidney capsule of normoglycaemic or hyperglycaemic, streptozotocin-induced, SCID mice and analyzed two weeks later. Vascularization was analyzed both quantitatively and qualitatively using either in situ hybridization with both mouse- and chick-specific RNA probes for VEGFR2 or immunohistochemistry with an antibody to nestin, a marker of endothelial cells that is specific for murine cells. To inhibit angiopoietin 2 (Ang2), SCID mice were treated with 4 mg/kg IP L1–10 twice/week. Results In normoglycaemic condition, chicken-derived endocrine and exocrine cells developed well and intragraft vessels were lined with mouse endothelial cells. When pancreases were grafted in hyperglycaemic mice, growth and differentiation of the graft were altered and we observed endothelial discontinuities, large blood-filled spaces. Vessel density was decreased. These major vascular anomalies were associated with strong over-expression of chick-Ang2. To explore the possibility that Ang2 over-expression could be a key step in vascular disorganization induced by hyperglycaemia, we treated mice with L1–10, an Ang-2 specific inhibitor. Inhibition of Ang2 improved vascularization and beta-cell density. Conclusions This work highlighted an important role of Ang2 in pancreatic vascular defects induced by hyperglycaemia.
Collapse
|
41
|
El-Khalawany M, Shaaban D, Sultan M, Abd Alsalam F. Inhibition of angiogenesis as a new therapeutic target in the treatment of lepromatous leprosy. Clin Cosmet Investig Dermatol 2011; 5:1-6. [PMID: 22291474 PMCID: PMC3267406 DOI: 10.2147/ccid.s26200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Angiogenesis was suggested to have a significant role in the pathogenesis of leprosy. However, the benefit of inhibiting angiogenesis in lepromatous leprosy patients has not previously been studied. The purpose of this study was to evaluate angiogenesis in leprosy patients before and after treatment with multidrug therapy (MDT) with and without minocycline. Methods A total of 40 patients with lepromatous leprosy were enrolled in this study. They were categorized into two equal groups (A and B), each formed of 20 patients. Group A received World Health Organization MDT, and Group B received MDT combined with minocycline, which has a known antiangiogenic effect. Microvascular density (MVD) in dermal granuloma was evaluated in both groups by immunostaining with CD31 and CD34 markers before and after 6 months of treatment. Results With CD31 immunostaining, the mean MVD in Group A significantly decreased from 39.1 ± 3.1 vessels (v)/high power field (HPF) to 16.5 ± 2.7 v/HPF, and in Group B it significantly decreased from 38.3 ± 2.5 v/HPF to 7.6 ± 1.9 v/HPF. CD34 immunostaining also showed a significant decrease of MVD from 42.2 ± 3.1 v/HPF to 18.8 ± 2.4 v/HPF in Group A, and in Group B it significantly decreased from 43.7 ± 2.3 v/HPF to 11.5 ± 1.6 v/HPF. The reduction of MVD was significantly higher in Group B compared with in Group A (P < 0.0001). Moreover, there was a significant reduction in bacterial density (assessed by bacterial index) in the cutaneous lesions of in Group B (decreased from 4.9 ± 0.3 to 1.4 ± 0.2) compared with in Group A (decreased from 5.1 ± 0.4 to 2.3 ± 0.4). Conclusion The synergistic effect of MDT and minocycline seems to be promising in the treatment of lepromatous leprosy. It significantly reduces angiogenesis and rapidly eliminates lepra bacilli from the skin that enables a rapid control and elimination of the disease.
Collapse
|
42
|
Kim Y, Kim BH, Lee H, Jeon B, Lee YS, Kwon MJ, Kim TY. Regulation of skin inflammation and angiogenesis by EC-SOD via HIF-1α and NF-κB pathways. Free Radic Biol Med 2011; 51:1985-95. [PMID: 21925591 DOI: 10.1016/j.freeradbiomed.2011.08.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 08/22/2011] [Accepted: 08/24/2011] [Indexed: 10/17/2022]
Abstract
Extracellular superoxide dismutase (EC-SOD) is an antioxidant enzyme that breaks down superoxide anion into oxygen and hydrogen peroxide in extracellular spaces and plays key roles in controlling pulmonary and vascular diseases in response to oxidative stresses. We aimed to investigate the role of EC-SOD in angiogenesis and inflammation in chronic inflammatory skin disorders such as psoriasis. Overexpressed EC-SOD reduced expression of angiogenic factors and proinflammatory mediators in hypoxia-induced keratinocytes and in ultraviolet B-irradiated mice, whereas the expression of the antiangiogenic factor tissue inhibitor of metalloproteinase-1 and anti-inflammatory cytokine interleukin-10 were increased. EC-SOD decreased new vessel formation, epidermal edema, and inflammatory cell infiltration in UVB-irradiated transgenic mice. Moreover, cells treated with recombinant human EC-SOD showed inhibited endothelial tube formation and cell proliferation. Overall, the antiangiogenic and anti-inflammatory effects of EC-SOD might be due to suppression of hypoxia-inducible factor-1α, protein kinase C, and nuclear factor-κB expression. Furthermore, EC-SOD expression in tissue from psoriasis patients was markedly decreased in psoriatic lesional and nonlesional skins from psoriasis patients in comparison to normal skin from healthy volunteers. Together, these results suggest that EC-SOD may provide a novel therapeutic approach to treating angiogenic and inflammatory skin diseases such as psoriasis.
Collapse
Affiliation(s)
- Younghwa Kim
- Department of Dermato-immunology, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Franklin MC, Navarro EC, Wang Y, Patel S, Singh P, Zhang Y, Persaud K, Bari A, Griffith H, Shen L, Balderes P, Kussie P. The structural basis for the function of two anti-VEGF receptor 2 antibodies. Structure 2011; 19:1097-107. [PMID: 21827946 DOI: 10.1016/j.str.2011.01.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 01/10/2011] [Accepted: 01/23/2011] [Indexed: 12/27/2022]
Abstract
The anti-VEGF receptor 2 antibody IMC-1121B is a promising antiangiogenic drug being tested for treatment of breast and gastric cancer. We have determined the structure of the 1121B Fab fragment in complex with domain 3 of VEGFR2, as well as the structure of a different neutralizing anti-VEGFR2 antibody, 6.64, also in complex with VEGFR2 domain 3. The two Fab fragments bind at opposite ends of VEGFR2 domain 3; 1121B directly blocks VEGF binding, whereas 6.64 may prevent receptor dimerization by perturbing the domain 3:domain 4 interface. Mutagenesis reveals that residues essential for VEGF, 1121B, and 6.64 binding are nonoverlapping among the three contact patches.
Collapse
|
44
|
Flisiak I, Zaniewski P, Rogalska-Taranta M, Chodynicka B. Effect of psoriasis therapy on VEGF and its soluble receptors serum concentrations. J Eur Acad Dermatol Venereol 2011; 26:302-7. [PMID: 21418334 DOI: 10.1111/j.1468-3083.2011.04053.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is recognized a pivotal pro-angiogenic factor responsible for new blood vessels formation in psoriatic lesion. OBJECTIVES The aim of the study was to analyse serum concentrations of VEGF and its soluble receptors (sVEGF) R1 and R2 in psoriatic patients before and after treatment. METHODS Serum samples were collected before and after 14 days of standard topical therapy, from 44 patients with exacerbated chronic plaque-type psoriasis and VEGF, sVEGF R1 and sVEGF R2 concentrations were measured using an enzyme immunoassay. Data were analysed with respect to baseline values of the psoriasis area severity index (PASI). RESULTS Baseline mean serum levels of VEGF and sVEGF R1, but not sVEGF R2 were significantly higher in patients than in healthy controls. VEGF demonstrated significant correlation with PASI score. Treatment resulted in significant reduction of VEGF serum concentration, particularly in patients with severe course of the disease (PASI >20) and increase in sVEGF R1 concentration in patients with mild disease activity (PASI <10). Moreover, serum sVEGF R1 level after treatment termination was significantly higher in patients with mild than severe course of psoriasis. CONCLUSIONS We confirmed the association between psoriasis activity and serum VEGF concentrations, which can be recognized as an indicator of the disease severity. However, the increase of serum sVEGF R1 concentrations can predict amelioration of clinical signs.
Collapse
Affiliation(s)
- I Flisiak
- Department of Dermatology and Venereology, Medical University of Białystok, Białystok, Poland.
| | | | | | | |
Collapse
|
45
|
Jung K, Lee D, Lim HS, Lee SI, Kim YJ, Lee GM, Kim SC, Koh GY. Double anti-angiogenic and anti-inflammatory protein Valpha targeting VEGF-A and TNF-alpha in retinopathy and psoriasis. J Biol Chem 2011; 286:14410-8. [PMID: 21345791 DOI: 10.1074/jbc.m111.228130] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pathological angiogenesis usually involves disrupted vascular integrity, vascular leakage, and infiltration of inflammatory cells, which are governed mainly by VEGF-A and TNF-α. Although many inhibitors targeting either VEGF-A or TNF-α have been developed, there is no single inhibitor molecule that simultaneously targets both molecules. Here, we designed and generated a novel chimeric decoy receptor (Valpha) that can simultaneously bind to VEGF-A and TNF-α and block their actions. In this experimental design, we have shown that Valpha, which is an effective synchronous blocker of VEGF-A and TNF-α, can drastically increase treatment effectiveness through its dual-blocking characteristics. Valpha contains the VEGF-A-binding domain of VEGFR1, the TNF-α-binding domain of TNFR2, and the Fc domain of IgG1. Valpha exhibited strong binding characteristics for its original counterparts, VEGF-A and TNF-α, but not for the extracellular matrix, resulting in a highly favorable pharmacokinetic profile in vivo. Compared with VEGF-Trap or Enbrel, both of which block either VEGF-A or TNF-α, singularly, Valpha is a highly effective molecule for reducing abnormal vascular tufts and the number of F4/80(+) macrophages in a retinopathy model. In addition, Valpha showed superior relief effects in a psoriasis model with regard to epidermal thickness and the area of blood and lymphatic vessels. Thus, the simultaneous blocking of VEGF-A and TNF-α using Valpha is an effective therapeutic strategy and cost-efficient for treatment of retinopathy and psoriasis.
Collapse
Affiliation(s)
- Keehoon Jung
- National Research Laboratory for Vascular Biology and Stem Cells, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Comparison of gene expression profiles reveals aberrant expression of FOXO1, Aurora A/B and EZH2 in lesional psoriatic skins. Mol Biol Rep 2010; 38:4219-24. [DOI: 10.1007/s11033-010-0544-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 11/16/2010] [Indexed: 12/15/2022]
|
47
|
Flisiak I, Zaniewski P, Rogalska M, Myśliwiec H, Jaroszewicz J, Chodynicka B. Effect of psoriasis activity on VEGF and its soluble receptors concentrations in serum and plaque scales. Cytokine 2010; 52:225-9. [PMID: 20980160 DOI: 10.1016/j.cyto.2010.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 09/26/2010] [Accepted: 09/30/2010] [Indexed: 02/06/2023]
Abstract
Vascular endothelial growth factor (VEGF) demonstrating pro-angiogenic activity promote new blood vessel formation in psoriatic lesions. The aim of this study was to evaluate the serum concentrations of VEGF, its soluble receptors (sVEGF R1 and R2) and VEGF content in scales of patients with psoriasis. To analyze possible association with activity of the disease, serum and scales from plaques were collected from 59 patients with exacerbated chronic plaque-type psoriasis. Mean concentrations of VEGF and sVEGF R1 in sera of patients were respectively two and four times higher than in healthy controls. Serum VEGF and sVEGF R1, but not sVEGF R2 demonstrated significant correlation with psoriasis area and severity index (PASI). There was also significant correlation between VEGF levels in serum and scales. Serum sVEGF R1 concentration was significantly elevated even in patients with low psoriasis activity (PASI<10), whereas increase of serum VEGF became significant in patients with medium activity (PASI: 10-20). Levels of serum VEGF and sVEGF R1 were the highest in patients with PASI>20. We confirmed association of both serum and scales VEGF concentrations with degree of psoriasis activity and demonstrated predominant increase of sVEGF R1 vs. VEGF in serum of patients with low psoriasis activity.
Collapse
Affiliation(s)
- Iwona Flisiak
- Department of Dermatology and Venereology Medical University of Bialystok, 15-540 Białystok, ul. Żurawia 14, Poland.
| | | | | | | | | | | |
Collapse
|
48
|
Hong I, Rho HS, Kim DH, Lee MO. Activation of LXRα induces lipogenesis in HaCaT cells. Arch Pharm Res 2010; 33:1443-9. [PMID: 20945144 DOI: 10.1007/s12272-010-0919-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 07/25/2010] [Accepted: 08/09/2010] [Indexed: 12/20/2022]
Abstract
The oxysterol nuclear receptors, LXRα (liver X receptor α; NR1H3) and LXRβ (NR1H2), coordinately regulate the expression of genes involved in lipid metabolism, anti-inflammation, and cholesterol transport. Previous studies have demonstrated that ligands of LXRα are important in the maintenance of the normal epidermal barrier function and keratinocyte differentiation. In this study, we examined whether LXRα and its ligands regulate lipid synthesis in HaCaT cells, a spontaneously transformed human keratinocyte cell line. When HaCaT cells were treated with the LXRα ligand TO901317, lipid droplets accumulated in the majority of cells, which were stained by Oil Red O. A luciferase reporter construct containing the LXR response element was activated about fourfold in HaCaT cells by TO901317 treatment, suggesting that LXR has a role in lipid synthesis in these cells. The expression of LXRα target genes, such as those encoding sterol regulatory binding protein and fatty acid synthase, were induced time dependently by TO901317, as measured by RT-PCR and western blotting. The expression of PPAR-α, -β, and -γ which regulate lipid metabolism, was also increased by TO901317 treatment. In contrast, TO901317 reduced the lipopolysaccharide-induced expression of cyclooxygenase 2 and inducible nitric oxide synthase in HaCaT cells. These results indicate that LXRα activation leads to lipogenesis in keratinocytes, which may enhance the epidermal barrier function of the skin.
Collapse
Affiliation(s)
- Il Hong
- College of Pharmacy and Bio-MAX Institute, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 151-742, Korea
| | | | | | | |
Collapse
|
49
|
Gilert A, Machluf M. Nano to micro delivery systems: targeting angiogenesis in brain tumors. JOURNAL OF ANGIOGENESIS RESEARCH 2010; 2:20. [PMID: 20932320 PMCID: PMC2964525 DOI: 10.1186/2040-2384-2-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 10/08/2010] [Indexed: 01/09/2023]
Abstract
Treating brain tumors using inhibitors of angiogenesis is extensively researched and tested in clinical trials. Although anti-angiogenic treatment holds a great potential for treating primary and secondary brain tumors, no clinical treatment is currently approved for brain tumor patients. One of the main hurdles in treating brain tumors is the blood brain barrier - a protective barrier of the brain, which prevents drugs from entering the brain parenchyma. As most therapeutics are excluded from the brain there is an urgent need to develop delivery platforms which will bypass such hurdles and enable the delivery of anti-angiogenic drugs into the tumor bed. Such delivery systems should be able to control release the drug or a combination of drugs at a therapeutic level for the desired time. In this mini-review we will discuss the latest improvements in nano and micro drug delivery platforms that were designed to deliver inhibitors of angiogenesis to the brain.
Collapse
Affiliation(s)
- Ariel Gilert
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| | | |
Collapse
|