1
|
Jang JH, Kim DH, Chun KS. Tumor microenvironment regulation by reactive oxygen species-mediated inflammasome activation. Arch Pharm Res 2025:10.1007/s12272-025-01532-6. [PMID: 39888519 DOI: 10.1007/s12272-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Tumor microenvironment (TME) is composed of diverse cell types whose interactions, both direct and indirect, significantly influence tumorigenesis and therapeutic outcomes. Within TME, reactive oxygen species (ROS) are produced by various cells and exhibit a dual role: moderate ROS levels promote tumor initiation and progression, whereas excessive levels induce cancer cell death, influencing the efficacy of anticancer therapies. Inflammasomes, cytosolic multiprotein complexes, are pivotal in multiple stages of tumorigenesis and play a crucial role in establishing the inflammatory TME. By releasing cytokines such as IL-1β and IL-18, inflammasomes contribute to immune cell recruitment and sustain a chronic inflammatory state that supports tumor growth. ROS are critical regulators of inflammasome activation, with the impact of ROS-mediated activation differing across cell types, leading to distinct influences on tumor progression and therapeutic responses. This review explores how ROS drive inflammasome activation in various TME-associated cells and the reciprocal ROS generation induced by inflammasomes, examining their multifaceted impact on tumorigenesis and therapeutic efficacy. By elucidating the complex interplay between ROS and inflammasomes in TME, we provide insights into potential therapeutic approaches that could modulate cancer progression and enhance treatment outcomes.
Collapse
Affiliation(s)
- Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan-si, Gyeongbuk, 38430, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
2
|
Jiang Z, Wang H, Wang X, Duo H, Tao Y, Li J, Li X, Liu J, Ni J, Wu EJ, Xiang H, Guan C, Wang X, Zhang K, Zhang P, Hou Z, Liu Y, Wang Z, Su B, Li B, Hao Y, Li B, Wu X. TMED4 facilitates regulatory T cell suppressive function via ROS homeostasis in tumor and autoimmune mouse models. J Clin Invest 2024; 135:e179874. [PMID: 39480507 PMCID: PMC11684806 DOI: 10.1172/jci179874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) plays crucial roles in maintaining Treg stability and function, yet the underlying mechanism remains largely unexplored. Here, we demonstrate that (Tmed4ΔTreg) mice with Treg-specific KO of ERS-related protein transmembrane p24 trafficking protein 4 (TMED4) had more Tregs with impaired Foxp3 stability, Treg signatures, and suppressive activity, which led to T cell hyperactivation and an exacerbated inflammatory phenotype and boosted antitumor immunity in mice. Mechanistically, loss of Tmed4 caused defects in ERS and a nuclear factor erythroid 2-related factor 2-related (NRF2-related) antioxidant response, which resulted in excessive ROS that reduced the Foxp3 stability and suppressive function of Tregs in an IRE1α/XBP1 axis-dependent manner. The abnormalities could be effectively rescued by the ROS scavenger, NRF2 inducer, or by forcible expression of IRE1α. Moreover, TMED4 suppressed IRE1α proteosome degradation via the ER-associated degradation (ERAD) system including the ER chaperone binding immunoglobulin protein (BIP). Our study reveals that TMED4 maintained the stability of Tregs and their suppressive function through IRE1α-dependent ROS and the NRF2-related antioxidant response.
Collapse
Affiliation(s)
- Zhenyan Jiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Huizi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xiaoxia Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
- Songjiang Research Institute, Songjiang District Central Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Hongrui Duo
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Yuexiao Tao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jia Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jiamin Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jun Ni
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Emily Jiatong Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Hongrui Xiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Chenyang Guan
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xinyu Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Kun Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Key Laboratory of Emotions and Affective Disorders, SJTU-SM, Shanghai, China
| | - Peng Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Key Laboratory of Emotions and Affective Disorders, SJTU-SM, Shanghai, China
| | - Zhaoyuan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, SJTU-SM, Shanghai, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, SJTU-SM, Shanghai, China
| | - Bing Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Bo Li
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Youjin Hao
- Research Group of Computational and Integrative Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xuefeng Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, and
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| |
Collapse
|
3
|
Ashraf M, El-Sawy HS, El Zaafarany GM, Abdel-Mottaleb MMA. Eucalyptus oil nanoemulsion for enhanced skin deposition of fluticasone propionate in psoriatic plaques: A combinatorial anti-inflammatory effect to suppress implicated cytokines. Arch Pharm (Weinheim) 2024:e2400557. [PMID: 39449230 DOI: 10.1002/ardp.202400557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects patients' quality of life. This study aimed to enhance the efficacy of topical application of fluticasone propionate (FP) using a eucalyptus oil-based nanoemulsion, an oil possessing anti-inflammatory activity and extracted from the leaves, fruits, and buds of Eucalyptus globulus or Eucalyptus maidenii, to improve the skin deposition of FP and aid its anti-inflammatory effect. Box-Behnken design was employed to optimize NE formulations, which were characterized for globule size, zeta potential, polydispersity index, rheological behavior, microscopic morphology, ex vivo skin permeation/deposition, and in vivo efficacy using imiquimod-induced psoriatic lesions. The optimized formulation depicted a droplet size of 188 ± 22.4 nm, a zeta potential of -17.63 ± 1.66 mV, and a viscosity of 204.9 mPa s. In addition to the increased FP retention in different skin layers caused by the NE and the reduced PASI score compared to the marketed cream, the levels of inflammatory cytokines IL-1α, IL-6, IL17a were markedly lowered, indicating the improved anti-psoriatic curable efficacy of the optimized formulation in comparison to the FP-marketed product.
Collapse
Affiliation(s)
- Mohamed Ashraf
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
- Department of Pharmacy, Al-Kut University College, Kut, Wasit, Iraq
| | - Ghada M El Zaafarany
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona M A Abdel-Mottaleb
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
4
|
Modestino L, Tumminelli M, Mormile I, Cristinziano L, Ventrici A, Trocchia M, Ferrara AL, Palestra F, Loffredo S, Marone G, Rossi FW, de Paulis A, Galdiero MR. Neutrophil exhaustion and impaired functionality in psoriatic arthritis patients. Front Immunol 2024; 15:1448560. [PMID: 39308858 PMCID: PMC11412820 DOI: 10.3389/fimmu.2024.1448560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Background Neutrophils (polymorphonuclear leukocytes, PMNs) are the most abundant subtype of white blood cells and are among the main actors in the inflammatory response. Psoriatic arthritis (PsA) is a chronic inflammatory disease affecting both the axial and peripheral joints. Typically associated with psoriasis, PsA can also affect multiple systems and organs, including the nails and entheses. Despite the involvement of PMNs in PsA, their specific role in the disease remains poorly understood. This study aimed to characterize the biological functions of PMNs and neutrophil-related mediators in PsA patients. Materials and methods 31 PsA patients and 22 healthy controls (HCs) were prospectively recruited. PMNs were isolated from peripheral blood and subjected to in vitro stimulation with lipopolysaccharide (LPS), N-Formylmethionyl-leucyl-phenylalanine (fMLP), tumor necrosis factor (TNF), phorbol 12-myristate 13-acetate (PMA), or control medium. Highly purified peripheral blood PMNs (>99%) were evaluated for activation status, reactive oxygen species (ROS) production, phagocytic activity, granular enzyme and neutrophil extracellular traps (NETs) release. Serum levels of matrix metalloproteinase-9 (MMP-9), myeloperoxidase (MPO), TNF, interleukin 23 (IL-23), and interleukin 17 (IL-17) were measured by ELISA. Serum Citrullinated histone H3 (CitH3) was measured as a NET biomarker. Results Activated PMNs from PsA patients displayed reduced activation, decreased ROS production, and impaired phagocytic activity upon stimulation with TNF, compared to HCs. PMNs from PsA patients also displayed reduced granular enzyme (MPO) and NET release. Serum analyses revealed elevated levels of MMP-9, MPO, TNF, IL-23, IL-17, and CitH3 in PsA patients compared to HCs. Serum CitH3 levels positively correlated with MPO and TNF concentrations, and IL-17 concentrations were positively correlated with IL-23 levels in PsA patients. These findings indicate that PMNs from PsA patients show reduced in vitro activation and function, and an increased presence of neutrophil-derived mediators (MMP-9, MPO, TNF, IL-23, IL-17, and CitH3) in their serum. Conclusions Taken together, our findings suggest that PMNs from PsA patients exhibit an "exhausted" phenotype, highlighting their plasticity and multifaceted roles in PsA pathophysiology.
Collapse
Affiliation(s)
- Luca Modestino
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
| | - Manuela Tumminelli
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
| | - Ilaria Mormile
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
| | - Leonardo Cristinziano
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Annagioia Ventrici
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
| | - Marialuisa Trocchia
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
| | - Francesco Palestra
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Institute of Experimental Endocrinology and Oncology ‘G. Salvatore’, National Research Council (CNR), Naples, Italy
| | - Francesca Wanda Rossi
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Amato de Paulis
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Internal Medicine and Clinical Immunology, University Hospital of Naples Federico II, Naples, Italy
- Department of Translational Medical Sciences (DiSMeT), University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Liu W, Jiang J, Li Z, Xiao Y, Zhou S, Wang D, Zou Y, Liu T, Li K, Liang H, Wang N, Xiang X, Xie Q, Zhan R, Zhang J, Zhou X, Yang L, Chuong CM, Lei M. Energy competition remodels the metabolic glucose landscape of psoriatic epidermal cells. Theranostics 2024; 14:3339-3357. [PMID: 38855186 PMCID: PMC11155411 DOI: 10.7150/thno.93764] [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: 12/31/2023] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Rationale: Skin cells actively metabolize nutrients to ensure cell proliferation and differentiation. Psoriasis is an immune-disorder-related skin disease with hyperproliferation in epidermal keratinocytes and is increasingly recognized to be associated with metabolic disturbance. However, the metabolic adaptations and underlying mechanisms of epidermal hyperproliferation in psoriatic skin remain largely unknown. Here, we explored the role of metabolic competition in epidermal cell proliferation and differentiation in psoriatic skin. Methods: Bulk- and single-cell RNA-sequencing, spatial transcriptomics, and glucose uptake experiments were used to analyze the metabolic differences in epidermal cells in psoriasis. Functional validation in vivo and in vitro was done using imiquimod-like mouse models and inflammatory organoid models. Results: We observed the highly proliferative basal cells in psoriasis act as the winners of the metabolic competition to uptake glucose from suprabasal cells. Using single-cell metabolic analysis, we found that the "winner cells" promote OXPHOS pathway upregulation by COX7B and lead to increased ROS through glucose metabolism, thereby promoting the hyperproliferation of basal cells in psoriasis. Also, to prevent toxic damage from ROS, basal cells activate the glutathione metabolic pathway to increase their antioxidant capacity to assist in psoriasis progression. We further found that COX7B promotes psoriasis development by modulating the activity of the PPAR signaling pathway by bulk RNA-seq analysis. We also observed glucose starvation and high expression of SLC7A11 that causes suprabasal cell disulfide stress and affects the actin cytoskeleton, leading to immature differentiation of suprabasal cells in psoriatic skin. Conclusion: Our study demonstrates the essential role of cellular metabolic competition for skin tissue homeostasis.
Collapse
Affiliation(s)
- Weiwei Liu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingwei Jiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zeming Li
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yang Xiao
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Siyi Zhou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Dehuan Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yi Zou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Tiantian Liu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Li
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Huan Liang
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Nian'ou Wang
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Xiao Xiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Qiaoli Xie
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Rixing Zhan
- State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Jinwei Zhang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Dermatology, Chongqing General Hospital, Chongqing 401147, China
| | - Xun Zhou
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Mingxing Lei
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
6
|
Nazimek K, Bryniarski K. Macrophage Functions in Psoriasis: Lessons from Mouse Models. Int J Mol Sci 2024; 25:5306. [PMID: 38791342 PMCID: PMC11121292 DOI: 10.3390/ijms25105306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Psoriasis is a systemic autoimmune/autoinflammatory disease that can be well studied in established mouse models. Skin-resident macrophages are classified into epidermal Langerhans cells and dermal macrophages and are involved in innate immunity, orchestration of adaptive immunity, and maintenance of tissue homeostasis due to their ability to constantly shift their phenotype and adapt to the current microenvironment. Consequently, both macrophage populations play dual roles in psoriasis. In some circumstances, pro-inflammatory activated macrophages and Langerhans cells trigger psoriatic inflammation, while in other cases their anti-inflammatory stimulation results in amelioration of the disease. These features make macrophages interesting candidates for modern therapeutic strategies. Owing to the significant progress in knowledge, our review article summarizes current achievements and indicates future research directions to better understand the function of macrophages in psoriasis.
Collapse
Affiliation(s)
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 31-121 Krakow, Poland;
| |
Collapse
|
7
|
Yu Y, Liu S, Yang L, Song P, Liu Z, Liu X, Yan X, Dong Q. Roles of reactive oxygen species in inflammation and cancer. MedComm (Beijing) 2024; 5:e519. [PMID: 38576456 PMCID: PMC10993368 DOI: 10.1002/mco2.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/21/2024] [Accepted: 02/23/2024] [Indexed: 04/06/2024] Open
Abstract
Reactive oxygen species (ROS) constitute a spectrum of oxygenic metabolites crucial in modulating pathological organism functions. Disruptions in ROS equilibrium span various diseases, and current insights suggest a dual role for ROS in tumorigenesis and the immune response within cancer. This review rigorously examines ROS production and its role in normal cells, elucidating the subsequent regulatory network in inflammation and cancer. Comprehensive synthesis details the documented impacts of ROS on diverse immune cells. Exploring the intricate relationship between ROS and cancer immunity, we highlight its influence on existing immunotherapies, including immune checkpoint blockade, chimeric antigen receptors, and cancer vaccines. Additionally, we underscore the promising prospects of utilizing ROS and targeting ROS modulators as novel immunotherapeutic interventions for cancer. This review discusses the complex interplay between ROS, inflammation, and tumorigenesis, emphasizing the multifaceted functions of ROS in both physiological and pathological conditions. It also underscores the potential implications of ROS in cancer immunotherapy and suggests future research directions, including the development of targeted therapies and precision oncology approaches. In summary, this review emphasizes the significance of understanding ROS-mediated mechanisms for advancing cancer therapy and developing personalized treatments.
Collapse
Affiliation(s)
- Yunfei Yu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Shengzhuo Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Luchen Yang
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Pan Song
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Zhenghuan Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Xiaoyang Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Xin Yan
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Qiang Dong
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
8
|
Cammarata I, Pinna V, Pacella I, Rotella I, Soresina A, Badolato R, Plebani A, Pignata C, Cirillo E, Zicari AM, Violi F, Carnevale R, Loffredo L, Piconese S. In adult X-CGD patients, regulatory T cells are expanded while activated T cells display a NOX2-independent ROS increase. Immunol Lett 2024; 266:106839. [PMID: 38309375 DOI: 10.1016/j.imlet.2024.106839] [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: 08/03/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
The X-linked chronic granulomatous disease (X-CGD), a rare genetic disease characterised by recurrent infections, is caused by mutations of NOX2. Significant proportions of X-CGD patients display signs of immune dysregulation. Regulatory T cells (Tregs) are CD4+T lymphocytes that expand in active inflammation and prevent autoimmune disorders. Here we asked whether X-CGD is associated to Treg dysfunctions in adult patients. To this aim, the frequency of Tregs was analysed through intracellular flow cytometry in a cohort of adult X-CGD patients, carriers and controls. We found that Tregs were significantly expanded and activated in blood of adult X-CGD patients, and this was associated with activation of conventional CD4+T cells (Tconvs). T cell activation was characterised by accumulation of intracellular ROS, not derived from NOX2 but likely produced by cellular metabolism. The higher TNF production by Tconvs in X-CGD patients might contribute to the expansion of Tregs through the TNFR2 receptor. In summary, our data indicate that Tregs expand in adult X-CGD in response to immune activation, and that the increase of NOX2-independent ROS content is a feature of activated T cells.
Collapse
Affiliation(s)
- Ilenia Cammarata
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Valeria Pinna
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilenia Pacella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ivano Rotella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Annarosa Soresina
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Raffaele Badolato
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Alessandro Plebani
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences - Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Emilia Cirillo
- Department of Translational Medical Sciences - Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; Mediterranea Cardiocentro-Napoli, Naples, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; IRCCS Neuromed, Località Camerelle, Pozzilli, Italy
| | - Lorenzo Loffredo
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy; Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
9
|
Bielecka E, Zubrzycka N, Marzec K, Maksylewicz A, Sochalska M, Kulawik-Pióro A, Lasoń E, Śliwa K, Malinowska M, Sikora E, Nowak K, Miastkowska M, Kantyka T. Ursolic Acid Formulations Effectively Induce Apoptosis and Limit Inflammation in the Psoriasis Models In Vitro. Biomedicines 2024; 12:732. [PMID: 38672088 PMCID: PMC11048670 DOI: 10.3390/biomedicines12040732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Psoriasis, a prevalent inflammatory skin disorder affecting a significant percentage of the global population, poses challenges in its management, necessitating the exploration of novel cost-effective and widely accessible therapeutic options. This study investigates the potential of ursolic acid (UA), a triterpenoid known for its anti-inflammatory and pro-apoptotic properties, in addressing psoriasis-related inflammation and keratinocyte hyperproliferation. The research involved in vitro models employing skin and immune cells to assess the effects of UA on psoriasis-associated inflammation. The presented research demonstrates the limiting effects of UA on IL-6 and IL-8 production in response to the inflammatory stimuli and limiting effects on the expression of psoriatic biomarkers S100A7, S100A8, and S100A9. Further, the study reveals promising outcomes, demonstrating UA's ability to mitigate inflammatory responses and hyperproliferation of keratinocytes by the induction of non-inflammatory apoptosis, as well as a lack of the negative influence on other cell types, including immune cells. Considering the limitations of UA's poor solubility, hybrid systems were designed to enhance its bioavailability and developed as hybrid nano-emulsion and bi-gel topical systems to enhance bioavailability and effectiveness of UA. One of them in particular-bi-gel-demonstrated high effectiveness in limiting the pathological response of keratinocytes to pro-psoriatic stimulation; this was even more prominent than with ursolic acid alone. Our results indicate that topical formulations of ursolic acid exhibit desirable anti-inflammatory activity in vitro and may be further employed for topical psoriasis treatment.
Collapse
Affiliation(s)
- Ewa Bielecka
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Cracow, Poland; (E.B.); (N.Z.); (K.M.); (A.M.)
| | - Natalia Zubrzycka
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Cracow, Poland; (E.B.); (N.Z.); (K.M.); (A.M.)
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland;
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland
| | - Karolina Marzec
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Cracow, Poland; (E.B.); (N.Z.); (K.M.); (A.M.)
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland;
| | - Anna Maksylewicz
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Cracow, Poland; (E.B.); (N.Z.); (K.M.); (A.M.)
| | - Maja Sochalska
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland;
| | - Agnieszka Kulawik-Pióro
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Elwira Lasoń
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Karolina Śliwa
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Magdalena Malinowska
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Elżbieta Sikora
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Krzysztof Nowak
- Wellnanopharm, Jerzego Samuela Bandtkego 19, 30-129 Cracow, Poland;
| | - Małgorzata Miastkowska
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland; (A.K.-P.); (E.L.); (K.Ś.); (M.M.); (E.S.); (M.M.)
| | - Tomasz Kantyka
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Cracow, Poland; (E.B.); (N.Z.); (K.M.); (A.M.)
| |
Collapse
|
10
|
Kondo M, Kumagai S, Nishikawa H. Metabolic advantages of regulatory T cells dictated by cancer cells. Int Immunol 2024; 36:75-86. [PMID: 37837615 DOI: 10.1093/intimm/dxad035] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 10/16/2023] Open
Abstract
Cancer cells employ glycolysis for their survival and growth (the "Warburg effect"). Consequently, surrounding cells including immune cells in the tumor microenvironment (TME) are exposed to hypoglycemic, hypoxic, and low pH circumstances. Since effector T cells depend on the glycolysis for their survival and functions, the metabolically harsh TME established by cancer cells is unfavorable, resulting in the impairment of effective antitumor immune responses. By contrast, immunosuppressive cells such as regulatory T (Treg) cells can infiltrate, proliferate, survive, and exert immunosuppressive functions in the metabolically harsh TME, indicating the different metabolic dependance between effector T cells and Treg cells. Indeed, some metabolites that are harmful for effector T cells can be utilized by Treg cells; lactic acid, a harmful metabolite for effector T cells, is available for Treg cell proliferation and functions. Deficiency of amino acids such as tryptophan and glutamine in the TME impairs effector T cell activation but increases Treg cell populations. Furthermore, hypoxia upregulates fatty acid oxidation via hypoxia-inducible factor 1α (HIF-1α) and promotes Treg cell migration. Adenosine is induced by the ectonucleotidases CD39 and CD73, which are strongly induced by HIF-1α, and reportedly accelerates Treg cell development by upregulating Foxp3 expression in T cells via A2AR-mediated signals. Therefore, this review focuses on the current views of the unique metabolism of Treg cells dictated by cancer cells. In addition, potential cancer combination therapies with immunotherapy and metabolic molecularly targeted reagents that modulate Treg cells in the TME are discussed to develop "immune metabolism-based precision medicine".
Collapse
Affiliation(s)
- Masaki Kondo
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shogo Kumagai
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
11
|
Ma Y, Sun T, Ren K, Min T, Xie X, Wang H, Xu G, Dang C, Zhang H. Applications of cold atmospheric plasma in immune-mediated inflammatory diseases via redox homeostasis: evidence and prospects. Heliyon 2023; 9:e22568. [PMID: 38107323 PMCID: PMC10724573 DOI: 10.1016/j.heliyon.2023.e22568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/28/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
As a representative technology in plasma medicine, cold atmospheric plasma (CAP) has beneficial outcomes in surface disinfection, wound repair, tissue regeneration, solid tumor therapy. Impact on immune response and inflammatory conditions was also observed in the process of CAP treatment. Relevant literatures were collected to assess efficacy and summarize possible mechanisms of the innovation. CAP mediates alteration in local immune microenvironment mainly through two ways. One is to down-regulate the expression level of several cytokines, impeding further conduction of immune or inflammatory signals. Intervening the functional phenotype of cells through different degree of oxidative stress is the other approach to manage the immune-mediated inflammatory disorders. A series of preclinical and clinical studies confirmed the therapeutic effect and side effects free of CAP. Moreover, several suggestions proposed in this manuscript might help to find directions for future investigation.
Collapse
Affiliation(s)
- Yuyi Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Tuanhe Sun
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Kaijie Ren
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Tianhao Min
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xin Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Haonan Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Guimin Xu
- State Key Laboratory of Electrical Insulation and Power Equipment, School of Electrical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
12
|
Trujillo-Ochoa JL, Kazemian M, Afzali B. The role of transcription factors in shaping regulatory T cell identity. Nat Rev Immunol 2023; 23:842-856. [PMID: 37336954 PMCID: PMC10893967 DOI: 10.1038/s41577-023-00893-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Forkhead box protein 3-expressing (FOXP3+) regulatory T cells (Treg cells) suppress conventional T cells and are essential for immunological tolerance. FOXP3, the master transcription factor of Treg cells, controls the expression of multiples genes to guide Treg cell differentiation and function. However, only a small fraction (<10%) of Treg cell-associated genes are directly bound by FOXP3, and FOXP3 alone is insufficient to fully specify the Treg cell programme, indicating a role for other accessory transcription factors operating upstream, downstream and/or concurrently with FOXP3 to direct Treg cell specification and specialized functions. Indeed, the heterogeneity of Treg cells can be at least partially attributed to differential expression of transcription factors that fine-tune their trafficking, survival and functional properties, some of which are niche-specific. In this Review, we discuss the emerging roles of accessory transcription factors in controlling Treg cell identity. We specifically focus on members of the basic helix-loop-helix family (AHR), basic leucine zipper family (BACH2, NFIL3 and BATF), CUT homeobox family (SATB1), zinc-finger domain family (BLIMP1, Ikaros and BCL-11B) and interferon regulatory factor family (IRF4), as well as lineage-defining transcription factors (T-bet, GATA3, RORγt and BCL-6). Understanding the imprinting of Treg cell identity and specialized function will be key to unravelling basic mechanisms of autoimmunity and identifying novel targets for drug development.
Collapse
Affiliation(s)
- Jorge L Trujillo-Ochoa
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
13
|
You M, Jiang Q, Huang H, Ma F, Zhou X. 4-Octyl itaconate inhibits inflammation to attenuate psoriasis as an agonist of oxeiptosis. Int Immunopharmacol 2023; 124:110915. [PMID: 37741130 DOI: 10.1016/j.intimp.2023.110915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023]
Abstract
Psoriasis is a highly prevalent chronic disease associated with a substantial social and economic burden. Oxeiptosis is a programmed cell death that occurs when cells are in a state of high oxidative stress, which has a potent anti-inflammatory effect. However, there is still no research on oxeiptosis in psoriasis, and the agonists or antagonists of oxeiptosis remain an unclear field. Here, we found that oxeiptosis of keratinocytes was inhibited in psoriasis lesions. KEAP1, as the upstream molecular component of oxeiptosis, is highly expressed in psoriasis lesions. Knockdown of KEAP1 in HaCaT cells caused oxeiptosis in the condition of M5 cocktail stimulation. Next, we found that the cell-permeable derivative of itaconate, 4-octylitaconate (OI) promoted oxeiptosis of keratinocytes by inhibiting KEAP1 and then activating PGAM5 which are two upstream molecular components of oxeiptosis. At the same time, OI can reduce the expression of inflammatory cytokines induced by M5 cocktail stimulation in vitro. Similarly, we found that OI can alleviate IMQ-induced psoriatic lesions in mice and downregulate the levels of inflammatory cytokines in psoriatic lesions. In summary, our findings suggest that oxeiptosis of keratinocytes was inhibited in psoriasis and OI can significantly inhibit inflammation and alleviate psoriasis as an agonist of oxeiptosis, indicating that oxeiptosis may be involved in regulating the progression of psoriasis, which may provide new therapeutic targets for psoriasis treatment.
Collapse
Affiliation(s)
- Mengshu You
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qian Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huining Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fangyu Ma
- Department of Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Xingchen Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
14
|
Sawase LR, Kumar TA, Mathew AB, Khodade VS, Toscano JP, Saini DK, Chakrapani H. β-Galactosidase-activated nitroxyl (HNO) donors provide insights into redox cross-talk in senescent cells. Chem Commun (Camb) 2023; 59:12751-12754. [PMID: 37811588 DOI: 10.1039/d3cc03094f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The cross-talk among reductive and oxidative species (redox cross-talk), especially those derived from sulfur, nitrogen and oxygen, influence several physiological processes including aging. One major hallmark of aging is cellular senescence, which is associated with chronic systemic inflammation. Here, we report a chemical tool that generates nitoxyl (HNO) upon activation by β-galactosidase, an enzyme that is over-expressed in senescent cells. In a radiation-induced senescence model, the HNO donor suppressed reactive oxygen species (ROS) in a hydrogen sulfide (H2S)-dependent manner. Hence, the newly developed tool provides insights into redox cross-talk and establishes the foundation for new interventions that modulate levels of these species to mitigate oxidative stress and inflammation.
Collapse
Affiliation(s)
- Laxman R Sawase
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Pune 411 008, Maharashtra, India.
| | - T Anand Kumar
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Pune 411 008, Maharashtra, India.
| | - Abraham B Mathew
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Deepak K Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Harinath Chakrapani
- Department of Chemistry, Indian Institute of Science Education and Research, Pune, Pune 411 008, Maharashtra, India.
| |
Collapse
|
15
|
Liu H, Luo Y, Zhao S, Tan J, Chen M, Liu X, Ye J, Cai S, Deng Y, Li J, He H, Zhang X, Zhong W. A reactive oxygen species-related signature to predict prognosis and aid immunotherapy in clear cell renal cell carcinoma. Front Oncol 2023; 13:1202151. [PMID: 37496661 PMCID: PMC10367095 DOI: 10.3389/fonc.2023.1202151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/09/2023] [Indexed: 07/28/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a malignant disease containing tumor-infiltrating lymphocytes. Reactive oxygen species (ROS) are present in the tumor microenvironment and are strongly associated with cancer development. Nevertheless, the role of ROS-related genes in ccRCC remains unclear. Methods We describe the expression patterns of ROS-related genes in ccRCC from The Cancer Genome Atlas and their alterations in genetics and transcription. An ROS-related gene signature was constructed and verified in three datasets and immunohistochemical staining (IHC) analysis. The immune characteristics of the two risk groups divided by the signature were clarified. The sensitivity to immunotherapy and targeted therapy was investigated. Results Our signature was constructed on the basis of glutamate-cysteine ligase modifier subunit (GCLM), interaction protein for cytohesin exchange factors 1 (ICEF1), methionine sulfoxide reductase A (MsrA), and strawberry notch homolog 2 (SBNO2) genes. More importantly, protein expression levels of GCLM, MsrA, and SBNO2 were detected by IHC in our own ccRCC samples. The high-risk group of patients with ccRCC suffered lower overall survival rates. As an independent predictor of prognosis, our signature exhibited a strong association with clinicopathological features. An accurate nomogram for improving the clinical applicability of our signature was constructed. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses showed that the signature was closely related to immune response, immune activation, and immune pathways. The comprehensive results revealed that the high-risk group was associated with high infiltration of regulatory T cells and CD8+ T cells and more benefited from targeted therapy. In addition, immunotherapy had better therapeutic effects in the high-risk group. Conclusion Our signature paved the way for assessing prognosis and developing more effective strategies of immunotherapy and targeted therapy in ccRCC.
Collapse
Affiliation(s)
- Hongxiang Liu
- School of Medicine, Jinan University, Guangzhou, China
- Department of Urology, The First People’s Hospital of Zhaoqing, Zhaoqing, China
| | - Yong Luo
- Department of Urology, The Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Shankun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Jing Tan
- Department of Pediatrics, The First People’s Hospital of Zhaoqing, Zhaoqing, China
| | - Minjian Chen
- Department of Urology, The First People’s Hospital of Zhaoqing, Zhaoqing, China
| | - Xihai Liu
- Department of Urology, The First People’s Hospital of Zhaoqing, Zhaoqing, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shanghua Cai
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou Laboratory, Guangzhou, China
| | - Yulin Deng
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jinchuang Li
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huichan He
- Guangzhou Medical University, Guangzhou Laboratory, Guangzhou, China
| | - Xin Zhang
- Department of Pathology, The Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Weide Zhong
- School of Medicine, Jinan University, Guangzhou, China
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou Laboratory, Guangzhou, China
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, Macao SAR, China
| |
Collapse
|
16
|
Costa S, Bevilacqua D, Caveggion E, Gasperini S, Zenaro E, Pettinella F, Donini M, Dusi S, Constantin G, Lonardi S, Vermi W, De Sanctis F, Ugel S, Cestari T, Abram CL, Lowell CA, Rodegher P, Tagliaro F, Girolomoni G, Cassatella MA, Scapini P. Neutrophils inhibit γδ T cell functions in the imiquimod-induced mouse model of psoriasis. Front Immunol 2022; 13:1049079. [DOI: 10.3389/fimmu.2022.1049079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
BackgroundPsoriasis is a chronic skin disease associated with deregulated interplays between immune cells and keratinocytes. Neutrophil accumulation in the skin is a histological feature that characterizes psoriasis. However, the role of neutrophils in psoriasis onset and development remains poorly understood.MethodsIn this study, we utilized the model of psoriasiform dermatitis, caused by the repeated topical application of an imiquimod containing cream, in neutrophil-depleted mice or in mice carrying impairment in neutrophil functions, including p47phox -/- mice (lacking a cytosolic subunit of the phagocyte nicotinamide adenine dinucleotide phosphate - NADPH - oxidase) and Sykfl/fl MRP8-cre+ mice (carrying the specific deletion of the Syk kinase in neutrophils only), to elucidate the specific contribution of neutrophils to psoriasis development.ResultsBy analyzing disease development/progression in neutrophil-depleted mice, we now report that neutrophils act as negative modulators of disease propagation and exacerbation by inhibiting gammadelta T cell effector functions via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-mediated reactive oxygen species (ROS) production. We also report that Syk functions as a crucial molecule in determining the outcome of neutrophil and γδ T cell interactions. Accordingly, we uncover that a selective impairment of Syk-dependent signaling in neutrophils is sufficient to reproduce the enhancement of skin inflammation and γδ T cell infiltration observed in neutrophil-depleted mice.ConclusionsOverall, our findings add new insights into the specific contribution of neutrophils to disease progression in the IMQ-induced mouse model of psoriasis, namely as negative regulatory cells.
Collapse
|
17
|
Hu J, Bian Q, Ma X, Xu Y, Gao J. A double-edged sword: ROS related therapies in the treatment of psoriasis. Asian J Pharm Sci 2022; 17:798-816. [PMID: 36600897 PMCID: PMC9800958 DOI: 10.1016/j.ajps.2022.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/27/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2022] Open
Abstract
In the onset and progression of psoriasis, redox imbalance is a vital factor. It's widely accepted that too much reactive oxygen species (ROS) always make psoriasis worse. Recent research, however, has shown that the accumulation of ROS is not entirely detrimental, as it helps reduce psoriasis lesions by inhibiting epidermal proliferation and keratinocyte death. As a result, ROS appears to have two opposing effects on the treatment of psoriasis. In this review, the current ROS-related therapies for psoriasis, including basic and clinical research, are presented. Additionally, the design and therapeutic benefits of various drug delivery systems and therapeutic approaches are examined, and a potential balance between anti-oxidative stress and ROS accumulation is also trying to be investigated.
Collapse
Affiliation(s)
- Jingyi Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiong Bian
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,College of Pharmacy, Inner Mongolia Medical University, Hohhot 010000, China
| | - Xiaolu Ma
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihua Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,Jiangsu Engineering Research Center for New-type External and Transdermal Preparations, Changzhou 213149, China,Corresponding author.
| |
Collapse
|
18
|
Lian J, Liang Y, Zhang H, Lan M, Ye Z, Lin B, Qiu X, Zeng J. The role of polyamine metabolism in remodeling immune responses and blocking therapy within the tumor immune microenvironment. Front Immunol 2022; 13:912279. [PMID: 36119047 PMCID: PMC9479087 DOI: 10.3389/fimmu.2022.912279] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The study of metabolism provides important information for understanding the biological basis of cancer cells and the defects of cancer treatment. Disorders of polyamine metabolism is a common metabolic change in cancer. With the deepening of understanding of polyamine metabolism, including molecular functions and changes in cancer, polyamine metabolism as a new anti-cancer strategy has become the focus of attention. There are many kinds of polyamine biosynthesis inhibitors and transport inhibitors, but not many drugs have been put into clinical application. Recent evidence shows that polyamine metabolism plays essential roles in remodeling the tumor immune microenvironment (TIME), particularly treatment of DFMO, an inhibitor of ODC, alters the immune cell population in the tumor microenvironment. Tumor immunosuppression is a major problem in cancer treatment. More and more studies have shown that the immunosuppressive effect of polyamines can help cancer cells to evade immune surveillance and promote tumor development and progression. Therefore, targeting polyamine metabolic pathways is expected to become a new avenue for immunotherapy for cancer.
Collapse
Affiliation(s)
- Jiachun Lian
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Hailiang Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Minsheng Lan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Ziyu Ye
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Bihua Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Xianxiu Qiu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jincheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
- Key Laboratory of Medical Bioactive Molecular Research for Department of Education of Guangdong Province, Collaborative Innovation Center for Antitumor Active Substance Research and Development, Zhanjiang, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
19
|
Zhang L, Liu X, Huang M, Wang R, Zhu W, Li Y, Shen L, Li C. Metformin Inhibits HaCaT Cell Proliferation Under Hyperlipidemia Through Reducing Reactive Oxygen Species via FOXO3 Activation. Clin Cosmet Investig Dermatol 2022; 15:1403-1413. [PMID: 35910506 PMCID: PMC9326038 DOI: 10.2147/ccid.s368845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022]
Abstract
Purpose Metformin (MET) has been proved to be effective for the treatment of psoriasis. The mechanisms of its action under the hyperlipidemia have yet to be fully elucidated. Here, we investigated the effect of metformin on the cell proliferation induced by hyperlipidemia and the underlying mechanism in immortalized human keratinocyte cell line (HaCat). Methods Wild-type or FOXO3 knockdown HaCat cells were treated with free fatty acids (FFA) for 10 days and then co-treated with metformin for another 4 days. Triglyceride (TG) level, cell viability, proliferation, apoptosis, antioxidant enzymes, reactive oxygen species (ROS) levels, as well as the transcription activity of FOXO3 were analyzed. Results Metformin decreased HaCaT cell proliferation and induced cell apoptosis after FFA treatment. Metformin was found to significantly increase the expressions and the activities of superoxide dismutase (SOD) as well as catalase (CAT), and reduced the reactive oxygen species (ROS) level. Metformin significantly promoted the autophagy and increase FOXO3 protein level in the nucleus under hyperlipidemia. However, all of the effects from metformin were partially blocked by FOXO3 knockdown. Conclusion This study demonstrated that under the hyperlipidemia, metformin has significant antiproliferation and proapoptosis effects by reducing ROS level as well as increasing autophagy. All of these effects from metformin were through FOXO3-dependent pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Xiaoling Liu
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Min Huang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Rui Wang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Wenwei Zhu
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Yu Li
- Department of Dermatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Lin Shen
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Chengxin Li
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| |
Collapse
|
20
|
Wang T, Xu H. Multi-faced roles of reactive oxygen species in anti-tumor T cell immune responses and combination immunotherapy. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
T cells play a central role in anti-tumor immunity, and reactive oxygen species (ROS) lie at the crossroad on the anti-tumor T cell responses. To activate efficient T cell immunity, a moderate level of ROS is needed, however, excessive ROS would cause toxicity to the T cells, because the improper level leads to the formation and maintenance of an immunosuppressive tumor microenvironment. Up to date, strategies that modulate ROS, either increasing or decreasing, have been widely investigated. Some of them are utilized in anti-tumor therapies, showing inevitable impacts on the anti-tumor T cell immunity with both obverse and reverse sides. Herein, the impacts of ROS-increasing and ROS-decreasing treatments on the T cell responses in the tumor microenvironment are reviewed and discussed. At the same time, outcomes of combination immunotherapies are introduced to put forward inspirations to unleash the potential of immunotherapies.
Collapse
Affiliation(s)
- Tao Wang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
21
|
So-In C, Sunthamala N. Treatment efficacy of Thunbergia laurifolia, Curcuma longa, Garcinia mangostana, and Andrographis paniculata extracts in Staphylococcus aureus-induced rabbit dermatitis model. Vet World 2022; 15:188-197. [PMID: 35369604 PMCID: PMC8924391 DOI: 10.14202/vetworld.2022.188-197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Dermatitis is a soft-tissue infection caused by Staphylococcus aureus. The recurrence of inflammatory skin is linked to clinical manifestations. Anti-inflammatory cytokines, which are essential for tissue damage, are released by bacteria through skin tissues. Oxidative stress causes inflammatory cells to necrotize and reduces their antioxidant profile, resulting in toxic damage to surrounding tissues. Although studies on the antibacterial effects of Thunbergia laurifolia Lindl., Curcuma longa L., Garcinia mangostana L., and Andrographis paniculata (Burm.). Bacterial infection of S. aureus have been conducted, most of these studies have been in vitro and were not related to the rabbit model. In addition, anti-inflammatory and antioxidant studies need to be evaluated. Thus, this study aims to compare the antibacterial, anti-inflammatory, and antioxidant properties of four local herbs with a standard antibiotic in S. aureus-induced rabbit dermatitis model. Materials and Methods: The skin of New Zealand white rabbits were artificially wounded using a sterile blade and then infected with S. aureus. The rabbits were divided into seven groups, each with three rabbits (Total 21 rabbits): The first group was the no infection group (no infection and no treatment with scarification), the second group was the no treatment group (S. aureus infection of the wound but no treatment), and the other five treated groups were T. laurifolia, C. longa, G. mangostana, A. paniculata, and bacitracin cream, all of which involved wound infection and treatments. The treatment lasted for 7 days. The antibacterial, anti-inflammatory, and antioxidant properties after treatment were measured. Results: The efficacy of T. laurifolia, C. longa, G. mangostana, and A. paniculata was similar to that of an antioxidant and free radical scavenging property. The bacterial infection process gradually reduced the activities of antioxidant systems (i.e., enzymatic levels and gene expressions) and total glutathione. However, the activities of the antioxidant system were steadily increased when treated with herbal extracts. During bacterial invasion of the skin, the concentration of thiobarbituric acid reactive molecules, the level of lipid peroxidation, and the expression of anti-inflammatory cytokine genes were increased. All these were decreased when herbal extracts were used to treat the lesion. Conclusion: It can be concluded that T. laurifolia, C. longa, G. mangostana, and A. paniculate extract have antibacterial, anti-inflammatory, and antioxidant properties and are effective antibacterial agents. G. mangostana is the most effective herbal extract for antidermatitis and has the potential to be used as an alternative topical treatment.
Collapse
Affiliation(s)
- Charinya So-In
- Department of Veterinary Technology, Faculty of Agricultural Technology, Kalasin University, Kalasin 46000, Thailand
| | - Nuchsupha Sunthamala
- Department of Biology, Faculty of Science, Mahasarakham University, Mahasarakham, 44150, Thailand
| |
Collapse
|
22
|
Elkoshi Z. Cancer and Autoimmune Diseases: A Tale of Two Immunological Opposites? Front Immunol 2022; 13:821598. [PMID: 35145524 PMCID: PMC8822211 DOI: 10.3389/fimmu.2022.821598] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 01/21/2023] Open
Abstract
The present article compares, side-by-side, cancer and autoimmune diseases in terms of innate and adaptive immune cells involvement, MHC Class I and Class II expression, TGFβ effect, immune modulating drugs effect and the effect of reactive oxygen species. The change in the inflammatory immune reaction during the progress of cancer and the effect of this change on the comorbidity of autoimmune diseases and cancer are discussed. The similar inflammatory properties of autoimmune diseases and early cancer, and the contrasting inflammatory properties of autoimmune diseases and advanced cancer elucidate the increased incidence of many types of cancer in patients with pre-existing autoimmune diseases and the decreased cancer-specific mortality of these patients. Stage-dependent effects of reactive oxygen-species on tumor proliferation are an additional probable cause for these epidemiological observations. The relationship: {standardized incidence ratio (SIR)} > {cancer-specific hazard ratio (HR)} for cancer patients with a history of autoimmune diseases is substantiated and rationalized.
Collapse
|
23
|
Shimojima Y, Kishida D, Ichikawa T, Takamatsu R, Nomura S, Sekijima Y. Oxidative Stress Promotes Instability of Regulatory T Cells in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Front Immunol 2021; 12:789740. [PMID: 34950150 PMCID: PMC8691772 DOI: 10.3389/fimmu.2021.789740] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/22/2021] [Indexed: 02/02/2023] Open
Abstract
We investigated the characteristics of regulatory T cells (Tregs), focusing on the relationship between their stability and reactive oxygen species (ROS), in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). Intracellular expressions of effector cytokines, forkhead box protein 3 (FoxP3), ROS, phosphorylated mammalian target of rapamycin (mTOR), and sirtuin 1 (SIRT1) in Tregs from peripheral blood mononuclear cells (PBMCs) of patients with AAV and healthy controls (HC) were analyzed. The alterations in and functional ability of Tregs were compared before and after resveratrol (RVL) treatment of PBMCs in patients with AAV. Significantly higher expressions of interferon (IFN)-γ, interleukin (IL)-17, IL-4, ROS, and phosphorylated mTOR (pho-mTOR) and lower expression of SIRT1 in CD4+CD25+FoxP3+ cells were found in patients with AAV than in the HC. FoxP3 expression in CD4+CD25+ cells and suppressive function of Tregs were significantly lower in patients with AAV than in the HC. Tregs after RVL treatment demonstrated significant decreases in IFN-γ, ROS, and pho-mTOR levels and increases in FoxP3, SIRT1 levels, and functional activity. Conversely, the direct activation of SIRT1 by SRT1720 resulted in decreased FoxP3 expression, with no reduction in ROS levels. The pho-mTOR levels were significantly higher in Tregs after activation by SRT1720 than in those after RVL treatment. This study suggested that imbalanced changes in Tregs could be attributed to mTOR activation, in which ROS overproduction was predominantly implicated. Therefore, ROS is a key mediator for promoting Tregs instability in AAV.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Shun Nomura
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
24
|
Wang J, Liu N, Jiang H, Li Q, Xing D. Reactive Oxygen Species in Anticancer Immunity: A Double-Edged Sword. Front Bioeng Biotechnol 2021; 9:784612. [PMID: 34869295 PMCID: PMC8635923 DOI: 10.3389/fbioe.2021.784612] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/29/2021] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) are critical mediators in many physiological processes including innate and adaptive immunity, making the modulation of ROS level a powerful strategy to augment anticancer immunity. However, current evidences suggest the necessity of a deeper understanding of their multiple roles, which may vary with their concentration, location and the immune microenvironment they are in. Here, we have reviewed the reported effects of ROS on macrophage polarization, immune checkpoint blocking (ICB) therapy, T cell activation and expansion, as well as the induction of immunogenic cell death. A majority of reports are indicating detrimental effects of ROS, but it is unadvisable to simply scavenge them because of their pleiotropic effects in most occasions (except in T cell activation and expansion where ROS are generally undesirable). Therefore, clinical success will need a clearer illustration of their multi-faced functions, as well as more advanced technologies to tune ROS level with high spatiotemporal control and species-specificity. With such progresses, the efficacy of current immunotherapies will be greatly improved by combining with ROS-targeted therapies.
Collapse
Affiliation(s)
- Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Ning Liu
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Qian Li
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao University, Qingdao, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
25
|
Mizuguchi S, Gotoh K, Nakashima Y, Setoyama D, Takata Y, Ohga S, Kang D. Mitochondrial Reactive Oxygen Species Are Essential for the Development of Psoriatic Inflammation. Front Immunol 2021; 12:714897. [PMID: 34421919 PMCID: PMC8378889 DOI: 10.3389/fimmu.2021.714897] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Psoriasis is a common immune-mediated, chronic, inflammatory skin disease that affects approximately 2-3% of the population worldwide. Although there is increasing evidence regarding the essential roles of the interleukin (IL)-23/IL-17 axis and dendritic cell (DC)-T cell crosstalk in the development of skin inflammation, the contributions of mitochondrial function to psoriasis are unclear. In a mouse model of imiquimod (IMQ)-induced psoriasiform skin inflammation, we found that hematopoietic cell-specific genetic deletion of p32/C1qbp, a regulator of mitochondrial protein synthesis and metabolism, protects mice from IMQ-induced psoriatic inflammation. Additionally, we demonstrate that p32/C1qbp is an important regulator of IMQ-induced DC activation, both in vivo and in vitro. We also found that p32/C1qbp-deficient DCs exhibited impaired production of IL-1β, IL-23, and mitochondrial reactive oxygen species (mtROS) after IMQ stimulation. Because the inhibition of mtROS suppressed IMQ-induced DC activation and psoriatic inflammation, we presume that p32/C1qbp and mtROS can serve as therapeutic targets in psoriasis.
Collapse
Affiliation(s)
- Soichi Mizuguchi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhito Gotoh
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Nakashima
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yurie Takata
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Sangaraju R, Alavala S, Nalban N, Jerald MK, Sistla R. Galangin ameliorates Imiquimod-Induced psoriasis-like skin inflammation in BALB/c mice via down regulating NF-κB and activation of Nrf2 signaling pathways. Int Immunopharmacol 2021; 96:107754. [PMID: 34162135 DOI: 10.1016/j.intimp.2021.107754] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022]
Abstract
Psoriasis is a most common chronic autoimmune-arbitrated cutaneous inflammatory skin disorder by unclear pathogenesis. In this current study we demonstrated the effect of galangin (GAL) on imiquimod (IMQ)-induced psoriasis-like skin inflammation and decipher its possible protective mechanism which has not been investigated. The in vivo results revealed that GAL at 1% w/w and 2% w/w for six consecutive days markedly reduced IMQ-induced PASI scoring, skin, ear thickness, hematological markers, levels of nitrites, TBARS, MPO, histopathological, as well modulated the protein levels of pro-inflammatory mediators of COX-2, iNOS, NF-κB pathway and pro-inflammatory cytokines IL-17, IL-23, IL-1β in the skin and also IL-6, TNF-α in both skin and serum. Besides, GAL restored the levels of antioxidants markers such as SOD, CAT, GST, GSH, GR and Vit-C, anti-inflammatory cytokine of IL-10, and the protein levels of Nrf2/HO-1 in the skin compared to the IMQ group. Finally, our study demonstrates that GAL exerted its protective effect by up-regulating the anti-inflammatory and the antioxidant markers against psoriasis pre-clinical models indicating its potency for treating psoriasis in humans.
Collapse
Affiliation(s)
- Rajendra Sangaraju
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Sateesh Alavala
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India
| | - Nasiruddin Nalban
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Mahesh Kumar Jerald
- Animal House Facility, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500 007, India
| | - Ramakrishna Sistla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
27
|
Horváth S, Kemény Á, Pintér E, Gyulai R. A Localized Aldara (5% Imiquimod)-Induced Psoriasiform Dermatitis Model in Mice Using Finn Chambers. ACTA ACUST UNITED AC 2021; 90:e78. [PMID: 32790966 DOI: 10.1002/cpph.78] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The expanding number of research studies utilizing the imiquimod-induced psoriasiform dermatitis model attests to the usefulness of this procedure. Advantages of this model include rapid development of the skin response and cost-effectiveness. A major limitation is that application of imiquimod cream over large areas of skin, as well as licking and ingestion of the cream, may lead to severe systemic inflammation, which can cause a general decline in health, splenomegaly, and death. In this protocol, Finn chambers are used to localize the imiquimod cream to a small area of the skin. This results in production of severe and reproducible psoriatic skin reactions with significantly less imiquimod, greatly reducing the possibility of untoward systemic effects. Moreover, having psoriasiform and control skin areas on the same mice decreases inter-animal differences. The protocol can be readily adapted for other skin disease models involving topical application of test agents. This article also details functional measurements performed during assays, including skin thickness, blood perfusion, semiquantitative histopathological evaluation, determination of scaling score to monitor psoriatic symptoms, and collection of spleen and body weight data to identify systemic effects. © 2020 The Authors. Basic Protocol: Use of Finn chambers to induce psoriasiform skin reactions with imiquimod Support Protocol 1: Measurement of double-fold dorsal skin thickness Support Protocol 2: Measurement of blood perfusion Support Protocol 3: Determination of scaling score Support Protocol 4: Semiquantitative histopathological scoring Support Protocol 5: Assessment of systemic side effects in response to imiquimod application.
Collapse
Affiliation(s)
- Szabina Horváth
- Department of Dermatology, Venereology and Oncodermatology, University of Pécs Clinical Center, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Ágnes Kemény
- János Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary.,Department of Medical Biology, University of Pécs Medical School, Pécs, Hungary
| | - Erika Pintér
- János Szentágothai Research Center, University of Pécs, Pécs, Hungary.,Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary
| | - Rolland Gyulai
- Department of Dermatology, Venereology and Oncodermatology, University of Pécs Clinical Center, Pécs, Hungary
| |
Collapse
|
28
|
Chang CH, Pauklin S. ROS and TGFβ: from pancreatic tumour growth to metastasis. J Exp Clin Cancer Res 2021; 40:152. [PMID: 33941245 PMCID: PMC8091747 DOI: 10.1186/s13046-021-01960-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor β (TGFβ) signalling pathway switches between anti-tumorigenic function at early stages of cancer formation and pro-tumorigenic effects at later stages promoting cancer metastasis. A similar contrasting role has been uncovered for reactive oxygen species (ROS) in pancreatic tumorigenesis. Down-regulation of ROS favours premalignant tumour development, while increasing ROS level in pancreatic ductal adenocarcinoma (PDAC) enhances metastasis. Given the functional resemblance, we propose that ROS-mediated processes converge with the spatial and temporal activation of TGFβ signalling and thereby differentially impact early tumour growth versus metastatic dissemination. TGFβ signalling and ROS could extensively orchestrate cellular processes and this concerted function can be utilized by cancer cells to facilitate their malignancy. In this article, we revisit the interplay of canonical and non-canonical TGFβ signalling with ROS throughout pancreatic tumorigenesis and metastasis. We also discuss recent insight that helps to understand their conflicting effects on different stages of tumour development. These considerations open new strategies in cancer therapeutics.
Collapse
Affiliation(s)
- Chao-Hui Chang
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, OX3 7LD, Oxford, UK
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, OX3 7LD, Oxford, UK.
| |
Collapse
|
29
|
Gangadevi V, Thatikonda S, Pooladanda V, Devabattula G, Godugu C. Selenium nanoparticles produce a beneficial effect in psoriasis by reducing epidermal hyperproliferation and inflammation. J Nanobiotechnology 2021; 19:101. [PMID: 33849555 PMCID: PMC8042708 DOI: 10.1186/s12951-021-00842-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 03/24/2021] [Indexed: 11/17/2022] Open
Abstract
Background Psoriasis is a chronic autoimmune skin disease characterized by hyperproliferation of keratinocytes. Wide treatment options used to treat psoriasis is associated with various adverse effects. To overcome this nanoformulation is prepared. Selenium is an essential trace element and plays major role in oxidation reduction system. Toxicity and stability limits the applications of selenium. Toxicity can be reduced and stabilized upon preparation into nanoparticles. Results Selenium nanoparticles (SeNPs) exhibit potent apoptosis through the generation of reactive oxygen species (ROS) with cell cycle arrest. SeNPs topical gel application produced significant attenuation of psoriatic severity with the abrogation of acanthosis and splenomegaly. SeNPs reduced the phosphorylation and expressions of MAPKs, STAT3, GSK-3β, Akt along with PCNA, Ki67, and cyclin-D1. Conclusion SeNPs inhibit various inflammation and proliferation mediated pathways and could be an ideal candidate for psoriasis therapy. Materials and methods SeNPs were characterized and various techniques were used to determine apoptosis and other molecular mechanisms. In vivo studies were performed by inducing psoriasis with imiquimod (IMQ). SeNPs were administered via topical route.
Graphic Abstract ![]()
Collapse
Affiliation(s)
- Vinod Gangadevi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Geetanjali Devabattula
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
30
|
Lin W, Shen P, Song Y, Huang Y, Tu S. Reactive Oxygen Species in Autoimmune Cells: Function, Differentiation, and Metabolism. Front Immunol 2021; 12:635021. [PMID: 33717180 PMCID: PMC7946999 DOI: 10.3389/fimmu.2021.635021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Accumulated reactive oxygen species (ROS) directly contribute to biomacromolecule damage and influence various inflammatory responses. Reactive oxygen species act as mediator between innate and adaptive immune cells, thereby influencing the antigen-presenting process that results in T cell activation. Evidence from patients with chronic granulomatous disease and mouse models support the function of ROS in preventing abnormal autoimmunity; for example, by supporting maintenance of macrophage efferocytosis and T helper 1/T helper 2 and T helper 17/ regulatory T cell balance. The failure of many anti-oxidation treatments indicates that ROS cannot be considered entirely harmful. Indeed, enhancement of ROS may sometimes be required. In a mouse model of rheumatoid arthritis (RA), absence of NOX2-derived ROS led to higher prevalence and more severe symptoms. In patients with RA, naïve CD4+ T cells exhibit inhibited glycolysis and enhanced pentose phosphate pathway (PPP) activity, leading to ROS exhaustion. In this "reductive" state, CD4+ T cell immune homeostasis is disrupted, triggering joint destruction, together with oxidative stress in the synovium.
Collapse
Affiliation(s)
- Weiji Lin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqin Song
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Saksida T, Jevtić B, Djedović N, Miljković Đ, Stojanović I. Redox Regulation of Tolerogenic Dendritic Cells and Regulatory T Cells in the Pathogenesis and Therapy of Autoimmunity. Antioxid Redox Signal 2021; 34:364-382. [PMID: 32458699 DOI: 10.1089/ars.2019.7999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Autoimmune diseases are progressively affecting westernized societies, as the proportion of individuals suffering from autoimmunity is steadily increasing over the past decades. Understanding the role of reactive oxygen species (ROS) in modulation of the immune response in the pathogenesis of autoimmune disorders is of utmost importance. The focus of this review is the regulation of ROS production within tolerogenic dendritic cells (tolDCs) and regulatory T (Treg) cells that have the essential role in the prevention of autoimmune diseases and significant potency in their therapy. Recent Advances: It is now clear that ROS are extremely important for the proper function of both DC and T cells. Antigen processing/presentation and the ability of DC to activate T cells depend upon the ROS availability. Treg differentiation, suppressive function, and stability are profoundly influenced by ROS presence. Critical Issues: Although a plethora of results on the relation between ROS and immune cells exist, it remains unclear whether ROS modulation is a productive way for skewing T cells and DCs toward a tolerogenic phenotype. Also, the possibility of ROS modulation for enhancement of regulatory properties of DC and Treg during their preparation for use in cellular therapy has to be clarified. Future Directions: Studies of DC and T cell redox regulation should allow for the improvement of the therapy of autoimmune diseases. This could be achieved through the direct therapeutic application of ROS modulators in autoimmunity, or indirectly through ROS-dependent enhancement of tolDC and Treg preparation for cell-based immunotherapy. Antioxid. Redox Signal. 34, 364-382.
Collapse
Affiliation(s)
- Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković," National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
32
|
Renin-Angiotensin System in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:105-114. [PMID: 33119868 DOI: 10.1007/978-3-030-50224-9_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
For enhancing the antitumor effects of current immunotherapies including immune-checkpoint blockade, it is important to reverse cancer-induced immunosuppression. The renin-angiotensin system (RAS) controls systemic body fluid circulation; however, the presence of a local RAS in tumors has been reported. Furthermore, the local RAS in tumors influences various immune and interstitial cells and affects tumor immune response. RAS stimulation through the angiotensin II type 1 receptor has been reported to inhibit tumor immune response. Therefore, RAS inhibitors and combined treatment with immunotherapy are expected in the future. In this chapter, we provide a background on the RAS and describe the tumor environment with regard to the RAS and tumor immune response.
Collapse
|
33
|
Mostafa A, Murata T, Kabashima K. Light in the dark: distinct effects of dimethyl fumarate on different T-cell subsets in psoriasis treatment. Br J Dermatol 2020; 184:389-390. [PMID: 32892342 DOI: 10.1111/bjd.19407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 11/30/2022]
Affiliation(s)
- A Mostafa
- Department of Dermatology, Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - T Murata
- Department of Dermatology, Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - K Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto, 606-8507, Japan.,A*STAR, Singapore Immunology Network (SIgN)/Skin Research Institute of Singapore (SRIS), Singapore
| |
Collapse
|
34
|
Kisipan ML, Ojoo RO, Kanui TI, Abelson KSP. Imiquimod does not elicit inflammatory responses in the skin of the naked mole rat (Heterocephalus glaber). BMC Res Notes 2020; 13:416. [PMID: 32891181 PMCID: PMC7487461 DOI: 10.1186/s13104-020-05260-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/27/2020] [Indexed: 11/29/2022] Open
Abstract
Objective Naked mole rat (Heterocephalus glaber) has recently attracted interest in biomedical research due to its exceptional longevity, cancer resistance and tolerance to potentially harmful conditions or stimuli. Given its unique attributes, this study was designed to characterize inflammatory skin reactions of this animal to topical application of imiquimod, a toll-like receptor 7 and 8 agonist that triggers psoriasis-like skin reaction. Results Imiquimod did not cause the expected psoriasis-like skin changes. There was no epidermal thickening and a straight epidermo-dermal boundary was maintained. There was no parakeratosis and the granular layer of epidermis was well formed. In the dermis, there was no leukocyte infiltration. This points to an exceptional nature of inflammatory/immune responses of this animal, but the mechanism could not be explained by our results. Naked mole rat could be a valuable negative model for studying psoriasis and other inflammatory skin conditions but as a prerequisite, there is need for further investigations to establish the mechanisms behind its lack of response to imiquimod.
Collapse
Affiliation(s)
- Mosiany Letura Kisipan
- Department of Veterinary Anatomy & Physiology, Egerton University, Egerton, P.O. Box 536, Nakuru, 20115, Kenya. .,Department of Experimental Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| | - Rodi Omondi Ojoo
- Department of Veterinary Anatomy & Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| | - Titus I Kanui
- Department of Agricultural Sciences, South Eastern Kenya University, P.O. Box 170, Kitui, 90200, Kenya
| | - Klas S P Abelson
- Department of Experimental Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| |
Collapse
|
35
|
Gawriluk TR, Simkin J, Hacker CK, Kimani JM, Kiama SG, Ezenwa VO, Seifert AW. Complex Tissue Regeneration in Mammals Is Associated With Reduced Inflammatory Cytokines and an Influx of T Cells. Front Immunol 2020; 11:1695. [PMID: 32849592 PMCID: PMC7427103 DOI: 10.3389/fimmu.2020.01695] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
While mammals tend to repair injuries, other adult vertebrates like salamanders and fish regenerate damaged tissue. One prominent hypothesis offered to explain an inability to regenerate complex tissue in mammals is a bias during healing toward strong adaptive immunity and inflammatory responses. Here we directly test this hypothesis by characterizing part of the immune response during regeneration in spiny mice (Acomys cahirinus and Acomys percivali) vs. fibrotic repair in Mus musculus. By directly quantifying cytokines during tissue healing, we found that fibrotic repair was associated with a greater release of pro-inflammatory cytokines (i.e., IL-6, CCL2, and CXCL1) during acute inflammation in the wound microenvironment. However, reducing inflammation via COX-2 inhibition was not sufficient to reduce fibrosis or induce a regenerative response, suggesting that inflammatory strength does not control how an injury heals. Although regeneration was associated with lower concentrations of many inflammatory markers, we measured a comparatively larger influx of T cells into regenerating ear tissue and detected a local increase in the T cell associated cytokines IL-12 and IL-17 during the proliferative phase of regeneration. Taken together, our data demonstrate that a strong adaptive immune response is not antagonistic to regeneration and that other mechanisms likely explain the distribution of regenerative ability in vertebrates.
Collapse
Affiliation(s)
- Thomas R. Gawriluk
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Jennifer Simkin
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Corin K. Hacker
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - John M. Kimani
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Stephen G. Kiama
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Vanessa O. Ezenwa
- Odum School of Ecology, University of Georgia, Athens, GA, United States
- Department of Infectious Disease, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, KY, United States
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
36
|
Cytotoxicity of Saikosaponin A targets HEKa cell through apoptosis induction by ROS accumulation and inflammation suppression via NF-κB pathway. Int Immunopharmacol 2020; 86:106751. [PMID: 32634696 DOI: 10.1016/j.intimp.2020.106751] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
Saikosaponin A (SSA) is a triterpenoid saponin extracted from oriental medicinal plant Radix bupleuri, possessing various biological functions such as anti-inflammatory, immune regulation and anti-virus. This study aimed to explore therapeutic effects of SSA on psoriasis in both vitro and vivo. Our results showed that SSA increased reactive oxygen species (ROS) generation, and decreased mitochondrial membrane potential (MMP) and M5-induced inflammatory cytokines levels in HEKa cells in a dose-dependent manner. In addition, SSA promoted apoptosis and suppressed phosphorylation of NF-κB in vitro, which were restored by the ROS scavenger N-acetylcysteine (NAC). In imiquimod (IMQ)-induced mice, gavage with SSA markedly decreased Psoriasis Area and Severity Index (PASI) score and ameliorated epidermal hyperplasia through inhibition of NF-κB and NLRP3 signaling pathway. In conclusion, our studies demonstrate that SSA induces apoptosis and suppresses inflammation in HEKa cells and ameliorates IMQ-induced psoriasis in mice, making it a therapeutic candidate for psoriasis.
Collapse
|
37
|
The Influence of Reactive Oxygen Species in the Immune System and Pathogenesis of Multiple Sclerosis. Autoimmune Dis 2020; 2020:5793817. [PMID: 32789026 PMCID: PMC7334772 DOI: 10.1155/2020/5793817] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/14/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
Multiple roles have been indicated for reactive oxygen species (ROS) in the immune system in recent years. ROS have been extensively studied due to their ability to damage DNA and other subcellular structures. Noticeably, they have been identified as a pivotal second messenger for T-cell receptor signaling and T-cell activation and participate in antigen cross-presentation and chemotaxis. As an agent with direct toxic effects on cells, ROS lead to the initiation of the autoimmune response. Moreover, ROS levels are regulated by antioxidant systems, which include enzymatic and nonenzymatic antioxidants. Enzymatic antioxidants include superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase. Nonenzymatic antioxidants contain vitamins C, A, and E, glutathione, and thioredoxin. Particularly, cellular antioxidant systems have important functions in maintaining the redox system homeostasis. This review will discuss the significant roles of ROS generation and antioxidant systems under normal conditions, in the immune system, and pathogenesis of multiple sclerosis.
Collapse
|
38
|
Gao J, Guo J, Nong Y, Mo W, Fang H, Mi J, Qi Q, Yang M. 18β-Glycyrrhetinic acid induces human HaCaT keratinocytes apoptosis through ROS-mediated PI3K-Akt signaling pathway and ameliorates IMQ-induced psoriasis-like skin lesions in mice. BMC Pharmacol Toxicol 2020; 21:41. [PMID: 32493482 PMCID: PMC7271483 DOI: 10.1186/s40360-020-00419-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Background Psoriasis is a chronic inflammatory skin disease affecting 2–3% of the population worldwide. Hyperproliferative keratinocytes were thought to be an amplifier of inflammatory response, thereby sustaining persistence of psoriasis lesions. Agents with the ability to inhibit keratinocyte proliferation or induce apoptosis are potentially useful for psoriasis treatment. 18β-Glycyrrhetinic acid (GA), an active metabolite of glycyrrhizin, exhibits diverse pharmacological activities, including anti-inflammatory, anti-bacteria and anti-proliferation. The current study aims to evaluate the effects of GA on the proliferation and apoptosis of human HaCaT keratinocytes in vitro and investigate the effects of GA on the skin lesions of imiquimod (IMQ)-induced psoriasis-like mouse model in vivo. Methods Cell viability was assayed by CCK-8. Flow cytometry was performed to measure apoptosis and reactive oxygen species (ROS), with Annexin V-FITC/PI detection kit and DCFH-DA probe respectively. Caspase 9/3 activities were measured using caspase activity assay kits. The protein levels of Akt and p-Akt were determined using Western blotting. IMQ was applied to induce psoriasis-like skin lesions in mice. The histological change in mouse skin lesions was detected using hematoxylin and eosin (H&E) staining. The severity of skin lesions was scored based on Psoriasis Area Severity Index (PASI). RT-PCR was employed to examine the relative expression of TNF-α, IL-22 and IL-17A in mouse skin lesions. Results GA decreased HaCaT keratinocytes viability and induced cell apoptosis in a dose-dependent manner. In the presence of GA, intracellular ROS levels were significantly elevated. NAC, a ROS inhibitor, attenuated GA-mediated HaCaT keratinocytes growth inhibition and apoptosis. In addition, GA treatment remarkably decreased p-Akt protein level, which could be restored partially when cells were co-treated with GA and NAC. LY294002 (a PI3K inhibitor) treatment significantly enhanced GA-mediated cytotoxicity. Moreover, GA ameliorated IMQ-induced psoriasis-like skin lesions in mice. Conclusions GA inhibits proliferation and induces apoptosis in HaCaT keratinocytes through ROS-mediated inhibition of PI3K-Akt signaling pathway, and ameliorates IMQ-induced psoriasis-like skin lesions in mice.
Collapse
Affiliation(s)
- Jintao Gao
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China.
| | - Junfan Guo
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Yuejuan Nong
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Wenfei Mo
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Huanan Fang
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Jing Mi
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Qi Qi
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| | - Mengjuan Yang
- College of Biotechnology, Guilin Medical University, Guilin, 541100, Guangxi, People's Republic of China
| |
Collapse
|
39
|
Kim HR, Choi EJ, Kie JH, Lee JH, Seoh JY. Deficiency of glutathione peroxidase-1 and catalase attenuated diet-induced obesity and associated metabolic disorders. Acta Diabetol 2020; 57:151-161. [PMID: 31372751 DOI: 10.1007/s00592-019-01388-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022]
Abstract
AIMS Oxidative stress has been considered to contribute to the development of obesity-related metabolic disorders including insulin resistance. To the contrary, deficiency of an anti-oxidizing enzyme, glutathione peroxidase (GPx)-1, was reported to enhance insulin signaling, suggesting that oxidative stress may inhibit the development of type 2 diabetes. However, the beneficial effects of the absence of GPx-1 in metabolic homeostasis, including body weight control, have not yet been clearly manifested. To clarify the relationship between oxidative stress and obesity-related metabolic disorders, we investigated another mouse deficient with both GPx-1 and catalase (Cat). METHODS C57BL/6J wild-type and GPx-1-/- × Cat-/- mice were fed with a high-fat diet (60% fat) or a normal chow diet for 16 weeks and were investigated for metabolic and histological studies. RESULTS Body weight gain was significantly reduced, and glucose metabolism as well as hepatic steatosis was obviously improved in the GPx-1-/- × Cat-/- mice. The serum levels of insulin and total cholesterol were also significantly lowered. For the underlying mechanism, inflammation was attenuated and expression of markers for fat browning was enhanced in the visceral white adipose tissues. CONCLUSIONS Oxidative stress due to deficiency of GPx-1 and Cat may improve obesity-related metabolic disorders through attenuation of inflammation and fat browning.
Collapse
Affiliation(s)
- Hyung-Ran Kim
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea
| | - Eun-Jeong Choi
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea
| | - Jeong-Hae Kie
- Department of Pathology, National Health Insurance Cooperation Ilsan Hospital, Koyang, Republic of Korea
| | - Joo-Ho Lee
- Department of Surgery, Ewha Womans University Graduate School of Medicine, Gangseo-Gu, Seoul, Republic of Korea.
- Mediplant Research Institute of Bioscience, Mapo-Gu, Seoul, Republic of Korea.
| | - Ju-Young Seoh
- Department of Microbiology, Ewha Womans University Graduate School of Medicine, Magokdongro 2-25, Gangseo-Gu, Seoul, 07804, Republic of Korea.
- Mediplant Research Institute of Bioscience, Mapo-Gu, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Thatikonda S, Pooladanda V, Sigalapalli DK, Godugu C. Piperlongumine regulates epigenetic modulation and alleviates psoriasis-like skin inflammation via inhibition of hyperproliferation and inflammation. Cell Death Dis 2020; 11:21. [PMID: 31924750 PMCID: PMC6954241 DOI: 10.1038/s41419-019-2212-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/27/2022]
Abstract
Psoriasis is an autoimmune skin disease, where chronic immune responses due to exaggerated cytokine signaling, abnormal differentiation, and evasion of keratinocytes apoptosis plays a crucial role in mediating abnormal keratinocytes hyperproliferation. From the therapeutic perspective, the molecules with strong anti-proliferative and anti-inflammatory properties could have tremendous relevance. In this study, we demonstrated that piperlongumine (PPL) treatment effectively abrogated the hyperproliferation and differentiation of keratinocytes by inducing ROS-mediated late apoptosis with loss of mitochondrial membrane potential. Besides, the arrest of cell cycle was found at Sub-G1 phase as a result of DNA fragmentation. Molecularly, inhibition of STAT3 and Akt signaling was observed with a decrease in proliferative markers such as PCNA, ki67, and Cyclin D1 along with anti-apoptotic Bcl-2 protein expression. Keratin 17 is a critical regulator of keratinocyte differentiation, and it was found to be downregulated with PPL significantly. Furthermore, prominent anti-inflammatory effects were observed by inhibition of lipopolysaccharide (LPS)/Imiquimod (IMQ)-induced p65 NF-κB signaling cascade and strongly inhibited the production of cytokine storm involved in psoriasis-like skin inflammation, thus led to the restoration of normal epidermal architecture with reduction of epidermal hyperplasia and splenomegaly. In addition, PPL epigenetically inhibited histone-modifying enzymes, which include histone deacetylases (HDACs) of class I (HDAC1-4) and class II (HDAC6) evaluated by immunoblotting and HDAC enzyme assay kit. In addition, our results show that PPL effectively inhibits the nuclear translocation of p65 and a histone modulator HDAC3, thus sequestered in the cytoplasm of macrophages. Furthermore, PPL effectively enhanced the protein-protein interactions of HDAC3 and p65 with IκBα, which was disrupted by LPS stimulation and were evaluated by Co-IP and molecular modeling. Collectively, our findings indicate that piperlongumine may serve as an anti-proliferative and anti-inflammatory agent and could serve as a potential therapeutic option in treating psoriasis.
Collapse
Affiliation(s)
- Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Dilep Kumar Sigalapalli
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
41
|
Gawriluk TR, Simkin J, Hacker CK, Kimani JM, Kiama SG, Ezenwa VO, Seifert AW. Complex Tissue Regeneration in Mammals Is Associated With Reduced Inflammatory Cytokines and an Influx of T Cells. Front Immunol 2020. [PMID: 32849592 DOI: 10.3389/fimmu.2020.01695/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023] Open
Abstract
While mammals tend to repair injuries, other adult vertebrates like salamanders and fish regenerate damaged tissue. One prominent hypothesis offered to explain an inability to regenerate complex tissue in mammals is a bias during healing toward strong adaptive immunity and inflammatory responses. Here we directly test this hypothesis by characterizing part of the immune response during regeneration in spiny mice (Acomys cahirinus and Acomys percivali) vs. fibrotic repair in Mus musculus. By directly quantifying cytokines during tissue healing, we found that fibrotic repair was associated with a greater release of pro-inflammatory cytokines (i.e., IL-6, CCL2, and CXCL1) during acute inflammation in the wound microenvironment. However, reducing inflammation via COX-2 inhibition was not sufficient to reduce fibrosis or induce a regenerative response, suggesting that inflammatory strength does not control how an injury heals. Although regeneration was associated with lower concentrations of many inflammatory markers, we measured a comparatively larger influx of T cells into regenerating ear tissue and detected a local increase in the T cell associated cytokines IL-12 and IL-17 during the proliferative phase of regeneration. Taken together, our data demonstrate that a strong adaptive immune response is not antagonistic to regeneration and that other mechanisms likely explain the distribution of regenerative ability in vertebrates.
Collapse
Affiliation(s)
- Thomas R Gawriluk
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Jennifer Simkin
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - Corin K Hacker
- Department of Biology, University of Kentucky, Lexington, KY, United States
| | - John M Kimani
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Stephen G Kiama
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| | - Vanessa O Ezenwa
- Odum School of Ecology, University of Georgia, Athens, GA, United States.,Department of Infectious Disease, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, United States.,Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
42
|
Thatikonda S, Pooladanda V, Godugu C. Repurposing an old drug for new use: Niclosamide in psoriasis-like skin inflammation. J Cell Physiol 2019; 235:5270-5283. [PMID: 31846070 DOI: 10.1002/jcp.29413] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/05/2019] [Indexed: 12/13/2022]
Abstract
Drug discovery is an onerous, extremely expensive, and time-consuming process. Instead, drug repurposing is an attractive strategy for exploiting novel indications for a drug beyond its original use. The untapped potential of drug repurposing compensates the barriers associated with the drug discovery pipeline. Psoriasis is an autoimmune skin disease, where hyperproliferation of keratinocytes and exaggerated immune responses are the important hallmarks of the disease. Extensive in vitro and preclinical research has demonstrated that niclosamide was found to exert potent anticancer and anti-inflammatory properties by targeting STAT3, p65 NF-κB, and NFATc-1 signaling paradigm with minimal host toxicity. From the disease perspective, the static intracellular molecular network in both cancer and psoriasis share overlapping pathological features in terms of hyperproliferation and chronic inflammation, which is mediated by the aforementioned signaling cascade. The plausible mechanistic relevance has prompted us to investigate the implementation of niclosamide for repositioning in psoriasis. Our in vitro and in vivo findings suggest that niclosamide inhibits keratinocytes hyperproliferation by reactive oxygen species-mediated apoptosis through the loss of mitochondrial membrane potential, cell cycle arrest at Sub G1 phase, and DNA fragmentation. Furthermore, niclosamide treatment resulted in abrogation of lipopolysaccharide-induced inflammatory cytokine levels in murine macrophages. Additionally, our results provided a preclinical rationale in imiquimod (IMQ)-induced BALB/c mouse model, where niclosamide diligently mitigated the IMQ-induced epidermal hyperplasia and inflammation by downregulating STAT3, p65 NF-κB, and NFATc-1 transcription factors along with Akt, Ki-67, and ICAM-1 protein expression.
Collapse
Affiliation(s)
- Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| |
Collapse
|
43
|
Sanghera C, Teh JJ, Pinato DJ. The systemic inflammatory response as a source of biomarkers and therapeutic targets in hepatocellular carcinoma. Liver Int 2019; 39:2008-2023. [PMID: 31433891 DOI: 10.1111/liv.14220] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 08/10/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) strongly relates to inflammation, with chronic up-regulation of pro-inflammatory mediators standing as a potential unifying mechanism that underscores the origin and progression of HCC independent of aetiology. Activation of the diverse pro-inflammatory mediators either within the tumour or its microenvironment is part of an active cross-talk between the progressive HCC and the host, which is known to influence clinical outcomes including recurrence after radical treatments and long-term survival. A number of clinical biomarkers to measure the severity of cancer-related inflammation are now available, most of which emerge from routine blood parameters including neutrophil, lymphocyte, platelet counts, as well as albuminaemia and C-reactive protein levels. In this review, we summarise the body of evidence supporting the biologic qualification of inflammation-based scores in HCC and review their potential in facilitating the prognostic assessment and treatment allocation in the individual patient. We also discuss the evidence to suggest modulation of tumour-promoting inflammation may act as a source of novel therapeutic strategies in liver cancer.
Collapse
Affiliation(s)
| | - Jhia J Teh
- Department of Medicine, Imperial College London, London, UK
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
44
|
Fatty Acid Metabolites Combine with Reduced β Oxidation to Activate Th17 Inflammation in Human Type 2 Diabetes. Cell Metab 2019; 30:447-461.e5. [PMID: 31378464 PMCID: PMC8506657 DOI: 10.1016/j.cmet.2019.07.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 02/16/2019] [Accepted: 07/10/2019] [Indexed: 12/30/2022]
Abstract
Mechanisms that regulate metabolites and downstream energy generation are key determinants of T cell cytokine production, but the processes underlying the Th17 profile that predicts the metabolic status of people with obesity are untested. Th17 function requires fatty acid uptake, and our new data show that blockade of CPT1A inhibits Th17-associated cytokine production by cells from people with type 2 diabetes (T2D). A low CACT:CPT1A ratio in immune cells from T2D subjects indicates altered mitochondrial function and coincides with the preference of these cells to generate ATP through glycolysis rather than fatty acid oxidation. However, glycolysis was not critical for Th17 cytokines. Instead, β oxidation blockade or CACT knockdown in T cells from lean subjects to mimic characteristics of T2D causes cells to utilize 16C-fatty acylcarnitine to support Th17 cytokines. These data show long-chain acylcarnitine combines with compromised β oxidation to promote disease-predictive inflammation in human T2D.
Collapse
|
45
|
Freag MS, Torky AS, Nasra MMA, Abdelmonsif DA, Abdallah OY. Liquid crystalline nanoreservoir releasing a highly skin-penetrating berberine oleate complex for psoriasis management. Nanomedicine (Lond) 2019; 14:931-954. [DOI: 10.2217/nnm-2018-0345] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: The current work highlighted preparation of highly penetrating liquid crystalline nanoparticulates (LCNPs) reservoir of a solubility modified berberine oleate (Brb-OL) complex for effective psoriasis management. Materials & methods: Brb-OL-loaded LCNPs (Brb-OL-LCNPs) were prepared using hydrotrope method. Results: The proposed Brb-OL-LCNPs showed a particle size of 137 ± 3.7 nm and negative ζ-potential (-38 ± -5.85 mV). Brb-OL-LCNPs showed a threefold increase in the drug accumulated within rat skin and around tenfold increase in the drug permeation compared with crude Brb. In vivo studies revealed that topical application of Brb-OL-LCNPs hydrogel significantly alleviated psoriasis symptoms and reduced the levels of psoriatic inflammatory cytokines. Conclusion: Formulating Brb-OL in the LCNPs controlled the release, retention and permeation of the drug across skin layers, which are of prime importance for psoriasis management.
Collapse
Affiliation(s)
- May S Freag
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| | - Abrar S Torky
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| | - Maha MA Nasra
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt
| |
Collapse
|
46
|
Horváth S, Komlódi R, Perkecz A, Pintér E, Gyulai R, Kemény Á. Methodological refinement of Aldara-induced psoriasiform dermatitis model in mice. Sci Rep 2019; 9:3685. [PMID: 30842501 PMCID: PMC6403245 DOI: 10.1038/s41598-019-39903-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/30/2019] [Indexed: 02/06/2023] Open
Abstract
Imiquimod (IMQ)-induced skin inflammation is currently the most widely accepted psoriasis animal model, however, it features several limitations. We have modified the IMQ-model to minimize its systemic effects towards effectively maintaining the characteristic skin reactions. The original protocol (OP) uses 62.5 mg Aldara cream (or vaseline) on the shaved back skin of mice for 4 days. In contrast, in our modified protocol (MP) 25 mg Aldara and vaseline are applied simultaneously in separate Finn chambers over the dorsal skin of mice. In both the OP and MP groups, histology showed unequivocal hallmarks of psoriasiform dermatitis. Additionally, skin scaling and blood perfusion values were similar. While Aldara elicited significantly increased skin thickness in the MP group, significant weight loss, spleen enlargement, increased inflammatory cytokine levels in plasma, and treatment related death were only observed in the OP group. Our new method reproduces psoriatic skin alterations highlighting considerably reduced systemic inflammatory reactions. Possessing psoriasiform and control skin areas on the same mouse also reduces inter-individual differences. Additionally, the new method permits prolonged IMQ treatment studies to mimic the chronic nature of psoriasis. Finally, our experimental approach may also be used in other mouse models, to prevent the undesired systemic effects of topically applied drugs.
Collapse
Affiliation(s)
- Szabina Horváth
- Department of Dermatology, Venereology and Oncodermatology, University of Pécs, H-7632, Pécs, Akác str. 1, Hungary
| | - Rita Komlódi
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, H-7624, Pécs, Szigeti str. 12, Hungary
| | - Anikó Perkecz
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, H-7624, Pécs, Szigeti str. 12, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, H-7624, Pécs, Szigeti str. 12, Hungary
- János Szentágothai Research Center, University of Pécs, H-7624, Pécs, Ifjúság str. 20, Hungary
| | - Rolland Gyulai
- Department of Dermatology, Venereology and Oncodermatology, University of Pécs, H-7632, Pécs, Akác str. 1, Hungary
| | - Ágnes Kemény
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Medical School, H-7624, Pécs, Szigeti str. 12, Hungary.
- János Szentágothai Research Center, University of Pécs, H-7624, Pécs, Ifjúság str. 20, Hungary.
- Department of Medical Biology, University of Pécs, Medical School, H-7624, Pécs, Szigeti str. 12, Hungary.
| |
Collapse
|
47
|
Gan L, Duan J, Zhang S, Liu X, Poorun D, Liu X, Lu X, Duan X, Liu D, Chen H. Cold atmospheric plasma ameliorates imiquimod-induced psoriasiform dermatitis in mice by mediating antiproliferative effects. Free Radic Res 2019; 53:269-280. [PMID: 30663913 DOI: 10.1080/10715762.2018.1564920] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Psoriasis is a chronic hyperproliferative skin disease characterised by excessive growth of keratinocytes. Indeed, inducing keratinocyte apoptosis is a key mechanism responsible for psoriatic plaques clearance following some important existing therapies, which display pro-oxidant activity. Cold atmospheric plasma (CAP), acting as a tuneable source of reactive oxygen and nitrogen species (RONS), can controllably transfer RONS to the cellular environment, deliver antiproliferative RONS concentrations and exert antiproliferative and proapoptotic effects. This study was undertaken to evaluate the therapeutic potential of CAP in psoriasis. We used cell models of psoriasis-like inflammation by adding lipopolysaccharide (LPS) or tumour necrosis factor alpha (TNF-α) to HaCaT keratinocytes. Indirect plasma, plasma-activated medium (PAM), was administered to HaCaT cells. Atmospheric pressure plasma jet (APPJ) was applied directly to imiquimod (IMQ)-induced psoriasiform dermatitis in mice. The results showed that PAM induced an increase in intracellular ROS and caused keratinocyte apoptosis. Moreover, cells under inflammation showed lesser viability and larger apoptosis rate. With repeated administration of APPJ, psoriasiform lesions showed ameliorated morphological manifestation and reduced epidermal proliferation. Overall, this study supports that CAP holds good potential in psoriasis treatment.
Collapse
Affiliation(s)
- Lu Gan
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jiangwei Duan
- b State Key Laboratory of Advanced Electromagnetic Engineering and Technology, School of Electrical and Electronic Engineering, Huazhong University of Science and Technology , Huazhong University of Science and Technology , Wuhan , China
| | - Song Zhang
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xin Liu
- b State Key Laboratory of Advanced Electromagnetic Engineering and Technology, School of Electrical and Electronic Engineering, Huazhong University of Science and Technology , Huazhong University of Science and Technology , Wuhan , China
| | - Devesh Poorun
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xinxin Liu
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Xinpei Lu
- b State Key Laboratory of Advanced Electromagnetic Engineering and Technology, School of Electrical and Electronic Engineering, Huazhong University of Science and Technology , Huazhong University of Science and Technology , Wuhan , China
| | - Xiaoru Duan
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Dawei Liu
- b State Key Laboratory of Advanced Electromagnetic Engineering and Technology, School of Electrical and Electronic Engineering, Huazhong University of Science and Technology , Huazhong University of Science and Technology , Wuhan , China
| | - Hongxiang Chen
- a Department of Dermatology , Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
48
|
Esmaeili B, Mansouri P, Doustimotlagh AH, Izad M. Redox imbalance and IL-17 responses in memory CD4 + T cells from patients with psoriasis. Scand J Immunol 2018; 89:e12730. [PMID: 30375024 DOI: 10.1111/sji.12730] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 01/21/2023]
Abstract
All stages of the inflammatory process involved in T cell-mediated chronic skin disorders like psoriasis are affected by redox imbalance. On the other hand, Th17 cells have a critical role in the pathogenesis of psoriasis. In this study, we evaluated redox status in memory CD4 + T cells and plasma of patients with psoriasis and its correlation with IL-17 response. To this end, memory T cells were isolated from 10 patients with psoriasis and 10 controls. Intracellular Glutathione (GSH), reactive oxygen species (ROS) and superoxide as well as IL-17 were measured using flow cytometry. Plasma total anti-oxidant capacity (TAC) was quantified by ferric reducing ability of plasma (FRAP) assay. The expression of catalase (CAT), superoxide dismutase 1(SOD1), superoxide dismutase 2 (SOD2), nuclear factor, erythroid 2 like 2 (NFE2L2) and cytochrome b-245 beta chain (CYBB) genes were analysed using real-time PCR. Our results showed an increased intracellular ROS production in memory CD4 + T cells of patients compared to controls, (P = 0.04). Furthermore, a significant decrease in expression of catalase gene was found in patients, (P = 0.02). However, no significant differences were observed for intracellular GSH, IL-17 and TAC levels between patients and controls. Also, no correlation was seen between the intracellular IL-17 level and intracellular ROS, GSH and catalase gene expression levels. Collectively, we found an increased ROS production in stimulated memory T cells of patients that could be due to reduced expression of catalase gene. However, it seems that these redox abnormalities have no relationship with IL-17 response in memory T cells.
Collapse
Affiliation(s)
- Behnaz Esmaeili
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Immunology, Asthma and Allergy Research Institute (IAARI), Tehran University of Medical Sciences, Tehran, Iran
| | - Parvin Mansouri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Izad
- Immunology Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.,Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Metformin Promotes HaCaT Cell Apoptosis through Generation of Reactive Oxygen Species via Raf-1-ERK1/2-Nrf2 Inactivation. Inflammation 2018; 41:948-958. [PMID: 29549478 DOI: 10.1007/s10753-018-0749-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although metformin (MET) may be useful for the treatment of psoriasis, the mechanisms underlying its method of action have yet to be fully elucidated. Here, the relationship between MET function and reactive oxygen species (ROS) levels and the underlying mechanism were explored in human immortalized keratinocyte cell line (HaCaT). HaCaT cells were incubated with MET at 0, 10, 20, 40, and 60 mM for 24 h. The cell viability was evaluated by the CCK-8 assay. The cell apoptosis rate and intracellular ROS levels were examined using flow cytometry. The protein expression and the phosphorylation levels of nuclear factor erythroid-derived 2 related factor 2 (Nrf2), Raf-1, and ERK1/2 were assessed by Western blot. The specific ROS scavenger N-acetyl-cysteine (NAC) and the specific Nrf2 agonist Oltipraz (OPZ) were used to analyze the effect of MET. MET decreased HaCaT cell proliferation and induced HaCaT cell apoptosis in a dose-dependent manner. MET was found to elevate intracellular ROS levels in a dose-dependent manner, while pretreatment with NAC attenuated these effects. MET inhibits the protein expression and the phosphorylation levels of Nrf2. The combination of OPZ and MET can significantly increase the cell viability, decrease the rate of apoptosis, and attenuate the intracellular ROS levels relative to MET alone. MET inhibits the protein expression and the phosphorylation levels of Raf-1 and ERK1/2. MET was found to attenuate Raf-1-ERK1/2 signaling in HaCaT cells to suppress the expression and phosphorylation levels of Nrf2, which contributed to the intracellular generation of ROS and the pro-apoptotic effects of MET.
Collapse
|
50
|
Feduska JM, Tse HM. The proinflammatory effects of macrophage-derived NADPH oxidase function in autoimmune diabetes. Free Radic Biol Med 2018; 125:81-89. [PMID: 29723665 DOI: 10.1016/j.freeradbiomed.2018.04.581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/22/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease culminating in the destruction of insulin-producing pancreatic β-cells. While ultimately a T cell-mediated disease, macrophages play an indispensable role in disease initiation and progression. Infiltrating macrophages generate an inflammatory environment by releasing NADPH oxidase-derived superoxide and proinflammatory cytokines. The synthesis of reactive oxygen species (ROS) is acknowledged as putative factors contributing to autoimmunity and β-cell damage in T1D. In addition to direct lysis, free radicals collectively participate in β-cell destruction by providing a redox-dependent third signal necessary for islet-reactive CD4 and CD8 T cell maturation and by inducing oxidative post-translational modifications of β-cell epitopes to further exacerbate autoimmune responses. This review will provide an overview of macrophage function and a synergistic cross-talk with redox biology that contributes to autoimmune dysregulation in T1D.
Collapse
Affiliation(s)
- Joseph M Feduska
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182, United States
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182, United States.
| |
Collapse
|