1
|
Kim DH, Lee H, Kim MY, Hwangbo H, Ji SY, Bang E, Hong SH, Kim GY, Leem SH, Ryu D, Cheong J, Choi YH. Particulate matter 2.5 stimulates pyroptosis and necroptosis via the p38 MAPK/Akt/NF-κB signaling pathway in human corneal epithelial cells. Toxicology 2025; 515:154138. [PMID: 40199452 DOI: 10.1016/j.tox.2025.154138] [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: 01/17/2025] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Particulate matter 2.5 (PM2.5) exposure poses significant health risks, particularly to the eyes. This study aimed to investigate the cytotoxic effects of PM2.5 on human corneal epithelial cells (HCECs) and to elucidate the mechanisms involved in pyroptosis and necroptosis. HCECs were exposed to PM2.5, and cytotoxicity, reactive oxygen species (ROS) levels, and the expression of pyroptosis- and necroptosis-related proteins were assessed. The roles of nuclear factor-kappa B (NF-κB) and nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome signaling pathways were also investigated. Exposure to PM2.5 caused a dose-dependent decrease in cell viability, accompanied by significant NLRP3 inflammasome activation, leading to pyroptosis and the release of pro-inflammatory cytokines. Enhanced ROS generation and mitochondrial dysfunction have also been observed, along with indicators of necroptosis, such as increased levels of mixed-lineage kinase domain-like proteins. Importantly, activation of the NF-κB signaling pathway was crucial for these responses. The suppression of p38 mitogen-activated protein kinase (MAPK) and activation of protein kinase B (Akt) using pharmacological modulators SB203580 and SC79, respectively, significantly reduced PM2.5-mediated cellular damage. These findings indicate that p38 MAPK inhibition and Akt activation are key regulatory mechanisms that help attenuate the deleterious effects of PM2.5 on HCECs. In conclusion, our findings offer new insights into the mechanisms by which PM2.5 induces pyroptosis and necroptosis in HCECs, especially by activating the NLRP3 inflammasome and NF-κB signaling pathways. The critical regulatory roles of p38 MAPK and Akt underscore their potential as therapeutic targets to alleviate PM-induced ocular damage.
Collapse
Affiliation(s)
- Da Hye Kim
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Integrated Biological Science, The Graduate School of Pusan National University, Busan 46241, Republic of Korea.
| | - Hyesook Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea.
| | - Min Yeong Kim
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| | - Hyun Hwangbo
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| | - Seon Yeong Ji
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| | - EunJin Bang
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| | - Su Hyun Hong
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| | - Gi Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| | - Sun-Hee Leem
- Department of Biomedical Sciences, College of Natural Sciences, Dong-A University, Busan 49315, Republic of Korea; Department of Health Sciences, Dong-A University, Busan 49315, Republic of Korea.
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - JaeHun Cheong
- Department of Integrated Biological Science, The Graduate School of Pusan National University, Busan 46241, Republic of Korea; Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea.
| | - Yung Hyun Choi
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti‑Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| |
Collapse
|
2
|
Singh DD. NLRP3 inflammasome: structure, mechanism, drug-induced organ toxicity, therapeutic strategies, and future perspectives. RSC Med Chem 2025:d5md00167f. [PMID: 40370650 PMCID: PMC12070810 DOI: 10.1039/d5md00167f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
Drug-induced toxicity is an important issue in clinical medicine, which typically results in organ dysfunction and adverse health consequences. The family of NOD-like receptors (NLRs) includes intracellular proteins involved in recognizing pathogens and triggering innate immune responses, including the activation of the NLRP3 inflammasome. The NLRP3 (nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3) inflammasome is a critical component for both innate and adaptive immune responses and has been implicated in various drug-induced toxicities, including hepatic, renal, and cardiovascular diseases. The unusual activation of the NLRP3 inflammasome causes the release of pro-inflammatory cytokines, such as IL-1β and IL-18, which can lead to more damage to tissues. Targeting NLRP3 inflammasome is a potential therapeutic endeavour for suppressing drug-induced toxicity. This review provides insights into the mechanism, drug-induced organ toxicity, therapeutic strategies, and prospective therapeutic approaches of the NLRP3 inflammasome and summarizes the developing therapies that target the inflammasome unit. This review has taken up one of the foremost endeavours in understanding and inhibiting the NLRP3 inflammasome as a means of generating safer pharmacological therapies.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur 303002 India +91 9450078260
| |
Collapse
|
3
|
Agrawal S, Narang S, Shahi Y, Mukherjee S. Inhibitors of inflammasome (NLRP3) signaling pathway as promising therapeutic candidates for oral cancer. Biochim Biophys Acta Gen Subj 2025; 1869:130800. [PMID: 40180112 DOI: 10.1016/j.bbagen.2025.130800] [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: 09/10/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Inflammasomes are complex protein assemblies responsible for regulating the development and release of proinflammatory cytokines like interleukin-1beta (IL-1β) and interleukin-18 (IL-18) against the intracellular triggers. Among these, the Nod-like receptor protein 3 (NLRP3) inflammasome stands out as the most extensively studied and well-characterized member, implicated in numerous pathological conditions. A systematic literature search was conducted on the PubMed such as PubMed, Scopus, Google Scholar database to identify peer-reviewed publications pertaining to the role of NLRP3 in oral cancer pathogenesis and its inhibitors for targeted therapy. Recent research highlights the emerging significance of the NLRP3 inflammasome in tumorigenesis, garnering attention as a potential target for anticancer therapies. This review delves into the involvement of NLRP3 in cancer development and progression, providing an in-depth overview of its activation (and inhibition) and its impact on oral cancer pathogenesis. The manuscript provides a detailed review of the natural and synthetic compounds inhibiting the NLRP3 signaling pathway, which might act as therapeutic lead molecules in oral cancer. This holds promise to overcome targeted and effective treatment options the development of novel drugs targeting the NLRP3 inflammasome-mediated mechanisms in oral cancer.
Collapse
Affiliation(s)
- Shreya Agrawal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Shatakshi Narang
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India
| | - Yadvendra Shahi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India; Ram Manohar Lohia Institute of Medical Sciences (RMLIMS), Lucknow, Uttar Pradesh, India
| | - Sayali Mukherjee
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Uttar Pradesh, India.
| |
Collapse
|
4
|
Montazeri-Khosh Z, Ebrahimpour A, Keshavarz M, Sheybani-Arani M, Samiei A. Combination therapies and other therapeutic approaches targeting the NLRP3 inflammasome and neuroinflammatory pathways: a promising approach for traumatic brain injury. Immunopharmacol Immunotoxicol 2025; 47:159-175. [PMID: 39762721 DOI: 10.1080/08923973.2024.2444956] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVES Traumatic brain injury (TBI) precipitates a neuroinflammatory cascade, with the NLRP3 inflammasome emerging as a critical mediator. This review scrutinizes the complex activation pathways of the NLRP3 inflammasome by underscoring the intricate interplay between calcium signaling, mitochondrial disturbances, redox imbalances, lysosomal integrity, and autophagy. It is hypothesized that a combination therapy approach-integrating NF-κB pathway inhibitors with NLRP3 inflammasome antagonists-holds the potential to synergistically dampen the inflammatory storm associated with TBI. METHODS A comprehensive analysis of literature detailing NLRP3 inflammasome activation pathways and therapeutic interventions was conducted. Empirical evidence supporting the concurrent administration of MCC950 and Rapamycin was reviewed to assess the efficacy of dual-action strategies compared to single-agent treatments. RESULTS Findings highlight potassium efflux and calcium signaling as novel targets for intervention, with cathepsin B inhibitors showing promise in mitigating neuroinflammation. Dual therapies, particularly MCC950 and Rapamycin, demonstrate enhanced efficacy in reducing neuroinflammation. Autophagy promotion, alongside NLRP3 inhibition, emerges as a complementary therapeutic avenue to reverse neuroinflammatory damage. CONCLUSION Combination therapies targeting the NLRP3 inflammasome and related pathways offer significant potential to enhance recovery in TBI patients. This review presents compelling evidence for the development of such strategies, marking a new frontier in neuroinflammatory research and therapeutic innovation.
Collapse
Affiliation(s)
- Zana Montazeri-Khosh
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ahmad Ebrahimpour
- Student Research Committee, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mina Keshavarz
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Afshin Samiei
- Tobacco and Health Research Center, Endocrinology and Metabolism Research Center, Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
5
|
Hajare AD, Dagar N, Gaikwad AB. Klotho antiaging protein: molecular mechanisms and therapeutic potential in diseases. MOLECULAR BIOMEDICINE 2025; 6:19. [PMID: 40119098 PMCID: PMC11928720 DOI: 10.1186/s43556-025-00253-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/18/2025] [Accepted: 02/19/2025] [Indexed: 03/24/2025] Open
Abstract
Klotho, initially introduced as an anti-aging protein, is expressed in the brain, pancreas, and most prominently in the kidney. The two forms of Klotho (membrane-bound and soluble form) have diverse pharmacological functions such as anti-inflammatory, anti-oxidative, anti-fibrotic, tumour-suppressive etc. The membrane-bound form plays a pivotal role in maintaining kidney homeostasis by regulating fibroblast growth factor 23 (FGF 23) signalling, vitamin D metabolism and phosphate balance. Klotho deficiency has been linked with significantly reduced protection against various kidney pathological phenotypes, including diabetic kidney disease (DKD), which is a major cause of chronic kidney disease leading to end-stage kidney disease. Owing to the pleiotropic actions of klotho, it has shown beneficial effects in DKD by tackling the complex pathophysiology and reducing kidney inflammation, oxidative stress, as well as fibrosis. Moreover, the protective effect of klotho extends beyond DKD in other pathological conditions, including cardiovascular diseases, alzheimer's disease, cancer, inflammatory bowel disease, and liver disease. Therefore, this review summarizes the relationship between Klotho expression and various diseases with a special emphasis on DKD, the distinct mechanisms and the potential of exogenous Klotho supplementation as a therapeutic strategy. Future research into exogenous Klotho could unravel novel treatment avenues for DKD and other diseases.
Collapse
Affiliation(s)
- Aditya Dipakrao Hajare
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Neha Dagar
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, 333031, India.
| |
Collapse
|
6
|
Romana-Souza B, Mendes-Oliveira V, Sampaio AL, de Almeida Cabral Monteiro HC, da Silva CO, de Souza Nogueira J, Lagente V, Porto LC, Carneiro S. The expression of NLR family pyrin domain containing receptor 1 in Brazilian psoriatic patients is associated with increased disease severity and pro-inflammatory cytokine release. Arch Dermatol Res 2025; 317:476. [PMID: 39987325 DOI: 10.1007/s00403-025-04029-w] [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: 12/27/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
This study aimed to investigate the gene and protein expression of key inflammasome mediators in venous blood and skin biopsy samples from Brazilian psoriatic patients treated at the Pedro Ernesto University Hospital (HUPE). Samples were collected from both psoriatic patients and healthy controls. Gene expression was evaluated using quantitative reverse transcription polymerase chain reaction (qRT‒PCR), while protein levels were assessed through immunohistochemistry and multiplex immunoassays. Compared with those from control individuals, blood and skin biopsy samples from psoriatic patients had significantly higher mRNA levels of nucleotide-binding domain leucine-rich repeat (NLR) family pyrin domain containing receptor 1 (NLRP1), caspase-1, interleukin-1β (IL-1β), and interleukin-18 (IL-18). Immunohistochemical analysis revealed elevated protein levels of NLRP1, caspase-1, IL-1β, and IL-18 in psoriatic skin biopsies. Multiplex immunoassays revealed increased plasma levels of interferon-γ (IFN-γ), IL-1β, interleukin-17 A (IL-17 A), and tumor necrosis factor-α (TNF-α) in psoriatic patients. A positive correlation was observed between NLRP1 expression, disease severity, and the protein levels of IL-1β and TNF-α. In conclusion, in Brazilian psoriatic patients treated at HUPE, both blood and skin biopsy samples show increased expression of the NLRP1 inflammasome and its downstream mediators. Furthermore, NLRP1 expression is positively correlated with disease severity and the release of T helper 1-type cytokines, highlighting its potential role in the pathogenesis and progression of psoriasis.
Collapse
Affiliation(s)
- Bruna Romana-Souza
- Department of Histology and Embryology, State University of Rio de Janeiro, 381 Marechal Rondon Avenue, Rio de Janeiro, 20950-003, Brazil.
| | - Victoria Mendes-Oliveira
- Histocompatibility and Cryopreservation Laboratory, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Luisa Sampaio
- Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Camila Oliveira da Silva
- Histocompatibility and Cryopreservation Laboratory, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jeane de Souza Nogueira
- Histocompatibility and Cryopreservation Laboratory, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vincent Lagente
- NuMeCan Institute (Nutrition, Metabolism and Cancer), Univ Rennes, INSERM, INRAE, CHU Rennes, Rennes, F-35000, France
| | - Luís Cristóvão Porto
- Histocompatibility and Cryopreservation Laboratory, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sueli Carneiro
- School of Medicine and University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Kasti A, Katsas K, Nikolaki MD, Triantafyllou K. The Role and the Regulation of NLRP3 Inflammasome in Irritable Bowel Syndrome: A Narrative Review. Microorganisms 2025; 13:171. [PMID: 39858939 PMCID: PMC11767632 DOI: 10.3390/microorganisms13010171] [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: 11/20/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic disorder of the gastrointestinal tract. Its pathogenesis involves multiple factors, including visceral hypersensitivity and immune activation. NLRP3 inflammasome is part of the nucleotide-binding oligomerization domain-like receptor (NLR) family, a crucial component of the innate immune system. Preclinical studies have demonstrated that inhibiting NLRP3 reduces visceral sensitivity and IBS symptoms, like abdominal pain, and diarrhea, suggesting that targeting the NLRP3 might represent a novel therapeutic approach for IBS. This review aims to assess the NLRP3 inhibitors (tranilast, β-hydroxybutyrate, Chang-Kang-fang, paeoniflorin, coptisine, BAY 11-7082, and Bifidobacterium longum), highlighting the signaling pathways, and their potential role in IBS symptoms management was assessed. Although premature, knowledge of the action of synthetic small molecules, phytochemicals, organic compounds, and probiotics might make NLRP3 a new therapeutic target in the quiver of physicians' therapeutic choices for IBS symptoms management.
Collapse
Affiliation(s)
- Arezina Kasti
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Konstantinos Katsas
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Maroulla D. Nikolaki
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, 2nd Department of Internal Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, 12462 Athens, Greece
| |
Collapse
|
8
|
Ji Z, Feng X, Han C, Li S, Wu B, Zhang X, Zhu S, Tong W, Xu W. The malic acid inhibiting inflammation in ankylosing spondylitis by interfering M1 macrophage polarization. Int Immunopharmacol 2025; 144:113653. [PMID: 39566386 DOI: 10.1016/j.intimp.2024.113653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Ankylosing spondylitis (AS) is a motor system immune disease with significant genetic characteristics, resulting in joint fusion, deformity, rigidity, seriously affecting the quality of life of patients. Inflammatory bowel disease (IBD), characterized by intestinal mucosal damage and inflammatory changes, the most common extra-articular manifestation of AS. Due to the limitations of the application of therapeutic drugs, it is urgent to look for new mechanisms and strategies to effectively inhibit AS inflammation is. The content of malic acid (MA) was significantly decreased in peripheral blood of AS patients, and it was significantly negatively correlated with C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR). MA dramatically alleviated spinal damage and intestinal inflammation in mouse models of AS induced by β-1, 3-glucan solution. Mechanically, MA suppressed the NF-κB pathway by inhibiting polarization of M1-type macrophages, thereby alleviating spinal and intestinal inflammation. From the perspective of material metabolism, this study explored the mechanism by which MA, an intermediate product of glucose metabolism, reducing M1 polarization of macrophages to inhibit AS inflammation, providing a reliable basis for the pathogenesis research and precise targeted treatment of AS in the later stage.
Collapse
Affiliation(s)
- Zhou Ji
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xinzhe Feng
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Changhao Han
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Shuo Li
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Bin Wu
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xuchao Zhang
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Shanbang Zhu
- Department of Orthopaedics, Nanjing Jinling Hospital, Medical School of Nanjing University, 305 Zhongshandonglu Road, Nanjing 210002, China
| | - Wenwen Tong
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Weidong Xu
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
9
|
Hu XM, Zheng S, Zhang Q, Wan X, Li J, Mao R, Yang R, Xiong K. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J 2024; 23:64-76. [PMID: 38125299 PMCID: PMC10730955 DOI: 10.1016/j.csbj.2023.11.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests that regulated cell death, such as pyroptosis, apoptosis, and necroptosis, is deeply involved in the pathogenesis of psoriasis. As a newly recognized form of systematic cell death, PANoptosis is involved in a variety of inflammatory disorders through amplifying inflammatory and immune cascades, but its role in psoriasis remains elusive. OBJECTIVES To reveal the role of PANoptosis in psoriasis for a potential therapeutic strategy. METHODS Multitranscriptomic analysis and experimental validation were used to identify PANoptosis signaling in psoriasis. RNA-seq and scRNA-seq analyses were performed to establish a PANoptosis-mediated immune response in psoriasis, which revealed hub genes through WGCNA and predicted disulfiram as a potential drug. The effect and mechanism of disulfiram were verified in imiquimod (IMQ)-induced psoriasis. RESULTS Here, we found a highlighted PANoptosis signature in psoriasis patients through multitranscriptomic analysis and experimental validation. Based on this, two distinct PANoptosis patterns (non/high) were identified, which were the options for clinical classification. The high-PANoptosis-related group had a higher response rate to immune cell infiltration (such as M1 macrophages and keratinocytes). Subsequently, WGCNA showed the hub genes (e.g., S100A12, CYCS, NOD2, STAT1, HSPA4, AIM2, MAPK7), which were significantly associated with clinical phenotype, PANoptosis signature, and identified immune response in psoriasis. Finally, we explored disulfiram (DSF) as a candidate drug for psoriasis through network pharmacology, which ameliorated IMQ-mediated psoriatic symptoms through antipyroptosis-mediated inflammation and enhanced apoptotic progression. By analyzing the specific ligand-receptor interaction pairs within and between cell lineages, we speculated that DSF might exert its effects by targeting keratinocytes directly or targeting M1 macrophages to downregulate the proliferation of keratinocytes. CONCLUSIONS PANoptosis with its mediated immune cell infiltration provides a roadmap for research on the pathogenesis and therapeutic strategies of psoriasis.
Collapse
Affiliation(s)
- Xi-min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Shengyuan Zheng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510000, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
10
|
Zou S, Han X, Luo S, Tan Q, Huang H, Yao Z, Hou W, Jie H, Wang J. Bay-117082 treats sepsis by inhibiting neutrophil extracellular traps (NETs) formation through down-regulating NLRP3/N-GSDMD. Int Immunopharmacol 2024; 141:112805. [PMID: 39146778 DOI: 10.1016/j.intimp.2024.112805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024]
Abstract
During the inflammatory storm of sepsis, a significant quantity of neutrophil extracellular traps (NETs) are generated, which act as a double-edged sword and not only impede the invasion of foreign microorganisms but also exacerbate organ damage. This study provides evidence that NETs can cause damage to alveolar epithelial cells in vitro. The sepsis model developed in this study showed a significant increase in NETs in the bronchoalveolar lavage fluid (BALF). The development of NETs has been shown to increase the lung inflammatory response and aggravate injury to alveolar epithelial cells. Bay-117082, a well-known NF-κB suppressor, is used to modulate inflammation. This analysis revealed that Bay-117082 efficiently reduced total protein concentration, myeloperoxidase activity, and inflammatory cytokines in BALF. Moreover, Bay-117082 inhibited the formation of NETs, which in turn prevented the activation of the pore-forming protein gasdermin D (GSDMD). In summary, these results indicated that excessive NET production during sepsis exacerbated the onset and progression of acute lung injury (ALI). Therefore, Bay-117082 could serve as a novel therapeutic approach for ameliorating sepsis-associated ALI.
Collapse
Affiliation(s)
- Shujing Zou
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Xinai Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Shugeng Luo
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Quanguang Tan
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Huiying Huang
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Zhoulanlan Yao
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Wenjing Hou
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Hongyu Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jinghong Wang
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Mahjabeen A, Hasan MZ, Rahman MT, Islam MA, Khan RT, Kaiser MS. Genetic insights into the connection between pulmonary TB and non-communicable diseases: An integrated analysis of shared genes and potential treatment targets. PLoS One 2024; 19:e0312072. [PMID: 39432502 PMCID: PMC11493268 DOI: 10.1371/journal.pone.0312072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Pulmonary Tuberculosis (PTB) is a significant global health issue due to its high incidence, drug resistance, contagious nature, and impact on people with compromised immune systems. As mentioned by the World Health Organization (WHO), TB is responsible for more global fatalities than any other infectious illness. On the other side, WHO also claims that noncommunicable diseases (NCDs) kill 41 million people yearly worldwide. In this regard, several studies suggest that PTB and NCDs are linked in various ways and that people with PTB are more likely to acquire NCDs. At the same time, NCDs can increase susceptibility to active TB infection. Furthermore, because of potential drug interactions and therapeutic challenges, treating individuals with both PTB and NCDs can be difficult. This study focuses on seven NCDs (lung cancer (LC), diabetes mellitus (DM), Parkinson's disease (PD), silicosis (SI), chronic kidney disease (CKD), cardiovascular disease (CVD), and rheumatoid arthritis (RA)) and rigorously presents the genetic relationship with PTB regarding shared genes and outlines possible treatment plans. OBJECTIVES BlueThis study aims to identify the drug components that can regulate abnormal gene expression in NCDs. The study will reveal hub genes, potential biomarkers, and drug components associated with hub genes through statistical measures. This will contribute to targeted therapeutic interventions. METHODS Numerous investigations, including protein-protein interaction (PPI), gene regulatory network (GRN), enrichment analysis, physical interaction, and protein-chemical interaction, have been carried out to demonstrate the genetic correlation between PTB and NCDs. During the study, nine shared genes such as TNF, IL10, NLRP3, IL18, IFNG, HMGB1, CXCL8, IL17A, and NFKB1 were discovered between TB and the above-mentioned NCDs, and five hub genes (NFKB1, TNF, CXCL8, NLRP3, and IL10) were selected based on degree values. RESULTS AND CONCLUSION In this study, we found that all of the hub genes are linked with the 10 drug components, and it was observed that aspirin CTD 00005447 was mostly associated with all the other hub genes. This bio-informatics study may help researchers better understand the cause of PTB and its relationship with NCDs, and eventually, this can lead to exploring effective treatment plans.
Collapse
Affiliation(s)
- Amira Mahjabeen
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Md. Zahid Hasan
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Md. Tanvir Rahman
- Department of Information and Communication Technology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Md. Aminul Islam
- Health Informatics Research Lab, Department of Computer Science and Engineering, Daffodil International University, Dhaka, Bangladesh
| | - Risala Tasin Khan
- Institute of Information Technology, Jahangirnagar University, Dhaka, Bangladesh
| | - M. Shamim Kaiser
- Institute of Information Technology, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
12
|
Li Y, He Y, Yang F, Liang R, Xu W, Li Y, Cheng J, Liang B, Tang M, Shi X, Zhuang J, Luo M, Li L, Zhang R, Liu H, Jie H, Li X, Han X, Sun E, Zhai Z. Gasdermin E-mediated keratinocyte pyroptosis participates in the pathogenesis of psoriasis by promoting skin inflammation. Br J Dermatol 2024; 191:385-396. [PMID: 38655652 DOI: 10.1093/bjd/ljae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 02/29/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Psoriasis is a common chronic inflammatory disease with an unclear aetiology. Keratinocytes in psoriasis are susceptible to exogenous triggers that induce inflammatory cell death. OBJECTIVES To investigate whether gasdermin E (GSDME)-mediated pyroptosis in keratinocytes contributes to the pathogenesis of psoriasis. METHODS Skin samples from patients with psoriasis and from healthy controls were collected to evaluate the expression of GSDME, cleaved caspase-3 and inflammatory factors. We then analysed the data series GSE41662 to further compare the expression of GSDME between lesional and nonlesional skin samples in those with psoriasis. In vivo, a caspase-3 inhibitor and GSDME-deficient mice (Gsdme-/-) were used to block caspase-3/GSDME activation in an imiquimod-induced psoriasis model. Skin inflammation, disease severity and pyroptosis-related proteins were analysed. In vitro, tumour necrosis factor (TNF)-α-induced caspase-3/GSDME-mediated pyroptosis in the HACAT cell line was explored. RESULTS Our analysis of the GSE41662 data series found that GSDME was upregulated in psoriasis lesions vs. normal skin. High levels of inflammatory cytokines such as interleukin (IL)-1β, IL-6 and TNF-α were also found in psoriasis lesions. In mice in the Gsdme-/- and caspase-3 inhibitor groups, the severity of skin inflammation was attenuated and GSDME and cleaved caspase-3 levels decreased after imiquimod treatment. Similarly, IL-1β, IL-6 and TNF-α expression was decreased in the Gsdme-/- and caspase-3 inhibitor groups. In vitro, TNF-α induced HACAT cell pyroptosis through caspase-3/GSDME pathway activation, which was suppressed by blocking caspase-3 or silencing Gsdme. CONCLUSIONS Our study provides a novel explanation of TNF-α/caspase-3/GSDME-mediated keratinocyte pyroptosis in the initiation and -acceleration of skin inflammation and the progression of psoriasis.
Collapse
Affiliation(s)
- Yingfei Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Fangyuan Yang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Rongmei Liang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wenchao Xu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yehao Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jingbo Cheng
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Baozhu Liang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ming Tang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xingliang Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jian Zhuang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Minshuang Luo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Liuying Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ruilin Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huijuan Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hongyu Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xing Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinai Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Clinical Immunology, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Chen GA, Chen Y, Zhang Y, Zheng S, Zhu L, Ding M. Silicone dressing combined with topical oxygen therapy alleviates incontinence-associated dermatitis via NF-κB p65/STAT1 signaling pathway. Skin Res Technol 2024; 30:e13888. [PMID: 39099447 PMCID: PMC11298708 DOI: 10.1111/srt.13888] [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: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Incontinence-associated dermatitis (IAD) is a tough problem in clinical settings, not only increasing the risk of complications like catheter-related urinary tract infections and pressure ulcers in elderly and critically ill patients, but also prolonging hospital stays, raising hospital costs, and possibly leading to medical disputes. This study is aimed to evaluate the therapeutic effect of silicone dressing combined with topical oxygen therapy on IAD in a rat model. METHODS An IAD rat model induced by synthetic urine with trypsin was established. Hematoxylin & eosin staining was carried out to examine skin histology. Using immunofluorescence, the microvessel density in the affected skin tissues was determined. ELISA was performed to measure the concentrations of inflammatory cytokines and angiogenic factors in serum. The mRNA expression of EGF, PDGF, and VEGF was detected via qRT-PCR. Western blotting was employed to determine NF-κB p65/STAT1 pathway-related protein levels. RESULTS Compared to single therapy, silicone dressing combined with topical oxygen therapy could significantly reduce the severity of IAD, improve skin histology, inhibit inflammation, and promote angiogenesis in IAD rat models. Additionally, the results showed that relatively speaking, the combined therapy suppressed the NF-κB p65/STAT1 signaling pathway more effectively. CONCLUSION These findings indicated that silicone dressing combined with topical oxygen therapy can alleviate IAD through promoting wound healing and inhibiting inflammation via NF-κB p65/STAT1 signaling pathway in a rat model, which provided a theoretical basis for the prevention and treatment of IAD in clinic.
Collapse
Affiliation(s)
- Guiyu an Chen
- Department of NursingSchool of Medicine, Jinhua PolytechnicJinhuaZhejiangChina
| | - Yingxun Chen
- Department of General MedicineJinhua Municipal Central HospitalJinhuaZhejiangChina
| | - Yan Zhang
- Department of NursingSchool of Medicine, Jinhua PolytechnicJinhuaZhejiangChina
| | - Shufeng Zheng
- Department of GastroenterologyJinhua People's HospitalJinhuaZhejiangChina
| | - Louying Zhu
- Jinhua Center of Laboratory AnimalsJinhua Municipal Food and Drug Inspection InstituteJinhuaZhejiangChina
| | - Mingxing Ding
- Medical Molecular Biology LaboratorySchool of Medicine, Jinhua PolytechnicJinhuaZhejiangChina
| |
Collapse
|
14
|
Cebani L, Mvubu NE. Can We Exploit Inflammasomes for Host-Directed Therapy in the Fight against Mycobacterium tuberculosis Infection? Int J Mol Sci 2024; 25:8196. [PMID: 39125766 PMCID: PMC11311975 DOI: 10.3390/ijms25158196] [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: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tb), is a major global health issue, with around 10 million new cases annually. Advances in TB immunology have improved our understanding of host signaling pathways, leading to innovative therapeutic strategies. Inflammasomes, protein complexes organized by cytosolic pattern recognition receptors (PRRs), play a crucial role in the immune response to M. tb by activating caspase 1, which matures proinflammatory cytokines IL1β and IL18. While inflammation is necessary to fight infection, excessive or dysregulated inflammation can cause tissue damage, highlighting the need for precise inflammasome regulation. Drug-resistant TB strains have spurred research into adjunctive host-directed therapies (HDTs) that target inflammasome pathways to control inflammation. Canonical and non-canonical inflammasome pathways can trigger excessive inflammation, leading to immune system exhaustion and M. tb spread. Novel HDT interventions can leverage precision medicine by tailoring treatments to individual inflammasome responses. Studies show that medicinal plant derivatives like silybin, andrographolide, and micheliolide and small molecules such as OLT1177, INF39, CY-09, JJ002, Ac-YVAD-cmk, TAK-242, and MCC950 can modulate inflammasome activation. Molecular tools like gene silencing and knockouts may also be used for severe TB cases. This review explores these strategies as potential adjunctive HDTs in fighting TB.
Collapse
Affiliation(s)
| | - Nontobeko E. Mvubu
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
15
|
Ando T, Abe Y, Yamaji K, Nishikomori R, Tamura N. A case of cryopyrin-associated periodic syndrome due to somatic mosaic mutation complicated with recurrent circinate erythematous psoriasis. Mod Rheumatol Case Rep 2024; 8:368-372. [PMID: 38036300 DOI: 10.1093/mrcr/rxad067] [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: 10/03/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Cryopyrin-associated periotic syndrome (CAPS) is a rare autoinflammatory disease caused by genetic variants in innate immunity genes. Autoinflammatory diseases, including CAPS, mediate proinflammatory cytokines such as interleukin (IL)-1 and IL-18 and result in severe systemic inflammation. A gain-of-function mutation in the NLR family pyrin domain-containing 3 (NLRP3) gene, which encodes the protein cryopyrin, was identified to be responsible for CAPS in 2001, and since then several additional pathogenic mutations have been found. Moreover, other phenotypes have been identified based on severity and symptomatology, including familial cold autoinflammatory syndrome, Muckle-Wells syndrome, and neonatal-onset multisystem inflammatory disease/chronic neurologic cutaneous articular syndrome. Prompt diagnosis of CAPS remains challenging, however, due to unspecific, extensive clinical signs, and delayed diagnosis and treatment targeting IL-1 lead to multiorgan damage. Another factor complicating diagnosis is the existence of somatic mosaic mutations in the NLRP3 gene in some cases, resulting in symptoms and clinical courses that are atypical. The frequency of somatic mosaic mutations in CAPS was estimated to be 19% in a systematic review. Psoriasis is a chronic inflammatory skin disease that affects ∼3% of the global population. Although no reports have shown complication between CAPS and psoriasis, these diseases have several similarities and potential relationships, for instance activation of T helper 17 cells in the dermis and increased NLRP3 gene expression in psoriatic skin compared with normal skin. Here, we report a case of CAPS due to a somatic mosaic mutation with recurrent circinate erythematous psoriasis.
Collapse
Affiliation(s)
- Taiki Ando
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryuta Nishikomori
- The Department of Pediatrics and Child Health, Kurume University School of Medicine, Fukuoka, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Kamel FZ, Hoseiny HAM, Shahawy AAE, Boghdadi G, Shahawy AAE. NLRP3 (rs10754558) gene polymorphism and tumor necrosis factor alpha as predictors for disease activity and response to methotrexate and adalimumab in psoriasis. BMC Immunol 2024; 25:40. [PMID: 38965465 PMCID: PMC11223426 DOI: 10.1186/s12865-024-00630-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Psoriasis has a global prevalence of 1-3%, with variations observed across different ethnic groups and geographical areas. Disease susceptibility and response to anti-tumor necrosis factor-α (TNFα) drugs suggest different genetic regulatory mechanisms which may include NLR family pyrin domain containing 3 (NLRP3) polymorphism. Evaluation of the NLRP3 gene polymorphism, the serum level of CRP and TNFα in psoriasis patients and assessment of the NLRP3 (rs10754558) gene polymorphism, CRP and TNFα with disease severity and their role as biomarkers for response to Methotrexate and Adalimumab in psoriasis. The study had a total of 75 patients diagnosed with psoriasis vulgaris, who were compared to a control group of 75 healthy individuals. RESULTS There was a highly significant difference in NLRP3 genotypes and alleles distribution between psoriasis patients and controls (P = 0.002,0.004). The heterozygote genotype GC (OR = 3.67,95%CI:1.75-7.68, P = 0.0006), was linked with increased risk of psoriasis. Additionally, The GC genotype was significantly associated with nonresponse to psoriasis therapy (OR = 11.7,95%CI:3.24-42.28, P = 0.0002). Regarding serum CRP and TNFα levels, there was a highly statistically significant difference between psoriasis patients and controls (P < 0.0001), and there was also a highly statistically significant difference between responders and non-responders in psoriasis patients regarding PASI 50 (P < 0.0001). CONCLUSIONS The NLRP3 (rs10754558) genotypes GC was associated with the severe form of psoriasis and with nonresponse to psoriasis medication. Therefore, NLRP3 (rs10754558) gene polymorphism is an important prognostic biomarker in psoriasis patients. The serum TNFα can be used as a predictor for response to therapy in psoriasis patients. More research for evaluation of role of the NLRP3 gene polymorphism in the genetic risks and treatment outcomes associated with psoriasis is still required.
Collapse
Affiliation(s)
- Fatma Z Kamel
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Aya A El Shahawy
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ghada Boghdadi
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- College of Medicine, Ibn Sina National College, Jeddah, Saudi Arabia
| | - Alia A El Shahawy
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
17
|
Sun Y, Li F, Liu Y, Qiao D, Yao X, Liu GS, Li D, Xiao C, Wang T, Chi W. Targeting inflammasomes and pyroptosis in retinal diseases-molecular mechanisms and future perspectives. Prog Retin Eye Res 2024; 101:101263. [PMID: 38657834 DOI: 10.1016/j.preteyeres.2024.101263] [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: 07/30/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Retinal diseases encompass various conditions associated with sight-threatening immune responses and are leading causes of blindness worldwide. These diseases include age-related macular degeneration, diabetic retinopathy, glaucoma and uveitis. Emerging evidence underscores the vital role of the innate immune response in retinal diseases, beyond the previously emphasized T-cell-driven processes of the adaptive immune system. In particular, pyroptosis, a newly discovered programmed cell death process involving inflammasome formation, has been implicated in the loss of membrane integrity and the release of inflammatory cytokines. Several disease-relevant animal models have provided evidence that the formation of inflammasomes and the induction of pyroptosis in innate immune cells contribute to inflammation in various retinal diseases. In this review article, we summarize current knowledge about the innate immune system and pyroptosis in retinal diseases. We also provide insights into translational targeting approaches, including novel drugs countering pyroptosis, to improve the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Fan Li
- Eye Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yunfei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dijie Qiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyu Yao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dequan Li
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Tao Wang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangming District, Shenzhen, 518132, China; School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao You'anMen Street, Beijing, 100069, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
18
|
Chen X, Deng G, Chen K, Chen Y, Ye W, Sun P. Targeting the NLRP3 inflammasome in psoriasis. Int J Dermatol 2024; 63:844-851. [PMID: 38345734 DOI: 10.1111/ijd.17073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 07/05/2024]
Abstract
The NLRP3 inflammasome, a complex consisting of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3, has emerged as a critical mediator of pathological inflammation and a significant therapeutic target for various inflammatory diseases. Psoriasis, a chronic inflammatory skin condition without a definitive cure, has shown promising results in animal models through the inhibition of the NLRP3 inflammasome. This review aims to explore the development of the NLRP3 inflammasome in psoriasis and the molecular mechanisms responsible for its inhibition by natural products and small molecules currently being developed for psoriasis treatment. Furthermore, we are examining clinical trials using agents that block the NLRP3 pathway for the treatment of psoriasis. This study is timely to provide a new perspective on managing psoriasis.
Collapse
Affiliation(s)
- Xiuhui Chen
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Guoliang Deng
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Kaifeng Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Chen
- Department of Pharmacy, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Weijun Ye
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
19
|
Chen H, Dong K, Ding J, Xia J, Qu F, Lan F, Liao H, Qian Y, Huang J, Xu Z, Gu Z, Shi B, Yu M, Cui X, Yu Y. CRISPR genome-wide screening identifies PAK1 as a critical driver of ARSI cross-resistance in prostate cancer progression. Cancer Lett 2024; 587:216725. [PMID: 38364963 DOI: 10.1016/j.canlet.2024.216725] [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: 10/23/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Next-generation androgen receptor signaling inhibitors (ARSIs), such as enzalutamide (Enza) and darolutamide (Daro), are initially effective for the treatment of advanced prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). However, patients often relapse and develop cross-resistance, which consequently makes drug resistance an inevitable cause of CRPC-related mortality. By conducting a comprehensive analysis of GEO datasets, CRISPR genome-wide screening results, ATAC-seq data, and RNA-seq data, we systemically identified PAK1 as a significant contributor to ARSI cross-resistance due to the activation of the PAK1/RELA/hnRNPA1/AR-V7 axis. Inhibition of PAK1 followed by suppression of NF-κB pathways and AR-V7 expression effectively overcomes ARSI cross-resistance. Our findings indicate that PAK1 represents a promising therapeutic target gene for the treatment of ARSI cross-resistant PCa patients in the clinic. STATEMENT OF SIGNIFICANCE: PAK1 drives ARSI cross-resistance in prostate cancer progression.
Collapse
Affiliation(s)
- Haojie Chen
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China; Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Keqin Dong
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China; Department of Urology, Chinese PLA General Hospital of Central Theater Command, Wuhan, 430064, China
| | - Jie Ding
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Jia Xia
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Fajun Qu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Fuying Lan
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Haihong Liao
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Yuhang Qian
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Zihan Xu
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Zhengqin Gu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Bowen Shi
- Department of Urology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Mingming Yu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xingang Cui
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Yongjiang Yu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
20
|
Chen X, Zhang P, Zhang Y, Wei M, Tian T, Zhu D, Guan Y, Wei W, Ma Y. The research progression of direct NLRP3 inhibitors to treat inflammatory disorders. Cell Immunol 2024; 397-398:104810. [PMID: 38324950 DOI: 10.1016/j.cellimm.2024.104810] [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: 11/04/2023] [Revised: 01/08/2024] [Accepted: 01/15/2024] [Indexed: 02/09/2024]
Abstract
The NLRP3 inflammasome represents a cytoplasmic multiprotein complex with the capability to recognize a wide range of pathogen-derived, environmental, and endogenous stress-related factors. Dysregulated activation of the NLRP3 inflammasome has been implicated in the development of various inflammasome-associated disorders, highlighting its significance as a pivotal target for the treatment of inflammatory diseases. Nonetheless, despite its clinical importance, there is currently a lack of specific drugs available for directly targeting the NLRP3 inflammasome. Several strategies have been explored to target different facets of the NLRP3 inflammasome, with interventions aimed at directly inhibiting NLRP3 demonstrating the most promising efficacy and safety profiles. In this review, we provide a summary of direct inhibitors targeting NLRP3, elucidating their inhibitory mechanisms, clinical trial phases, and potential applications. Through this discussion, we aim to shed light on the implications of NLRP3 inhibition for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xiu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Pingping Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Yu Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Mengzhu Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Tian Tian
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Dacheng Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Yanling Guan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| | - Yang Ma
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammasome and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammasome and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
21
|
Kodi T, Sankhe R, Gopinathan A, Nandakumar K, Kishore A. New Insights on NLRP3 Inflammasome: Mechanisms of Activation, Inhibition, and Epigenetic Regulation. J Neuroimmune Pharmacol 2024; 19:7. [PMID: 38421496 PMCID: PMC10904444 DOI: 10.1007/s11481-024-10101-5] [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: 03/07/2023] [Accepted: 11/06/2023] [Indexed: 03/02/2024]
Abstract
Inflammasomes are important modulators of inflammation. Dysregulation of inflammasomes can enhance vulnerability to conditions such as neurodegenerative diseases, autoinflammatory diseases, and metabolic disorders. Among various inflammasomes, Nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) is the best-characterized inflammasome related to inflammatory and neurodegenerative diseases. NLRP3 is an intracellular sensor that recognizes pathogen-associated molecular patterns and damage-associated patterns resulting in the assembly and activation of NLRP3 inflammasome. The NLRP3 inflammasome includes sensor NLRP3, adaptor apoptosis-associated speck-like protein (ASC), and effector cysteine protease procaspase-1 that plays an imperative role in caspase-1 stimulation which further initiates a secondary inflammatory response. Regulation of NLRP3 inflammasome ameliorates NLRP3-mediated diseases. Much effort has been invested in studying the activation, and exploration of specific inhibitors and epigenetic mechanisms controlling NLRP3 inflammasome. This review gives an overview of the established NLRP3 inflammasome assembly, its brief molecular mechanistic activations as well as a current update on specific and non-specific NLRP3 inhibitors that could be used in NLRP3-mediated diseases. We also focused on the recently discovered epigenetic mechanisms mediated by DNA methylation, histone alterations, and microRNAs in regulating the activation and expression of NLRP3 inflammasome, which has resulted in a novel method of gaining insight into the mechanisms that modulate NLRP3 inflammasome activity and introducing potential therapeutic strategies for CNS disorders.
Collapse
Affiliation(s)
- Triveni Kodi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Adarsh Gopinathan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
22
|
Bu J, Mahan Y, Zhang S, Wu X, Zhang X, Zhou L, Zhang Y. Acacetin inhibits inflammation by blocking MAPK/NF-κB pathways and NLRP3 inflammasome activation. Front Pharmacol 2024; 15:1286546. [PMID: 38389927 PMCID: PMC10883387 DOI: 10.3389/fphar.2024.1286546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Objective: Our preliminary research indicates that acacetin modulates the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome, providing protection against Alzheimer's Disease (AD) and cerebral ischemic reperfusion injury. The mechanisms of acacetin to inhibit the activation of the NLRP3 inflammasome remain fully elucidated. This study aims to investigate the effects and potential mechanisms of acacetin on various agonists induced NLRP3 inflammasome activation. Methods: A model for the NLRP3 inflammasome activation was established in mouse bone marrow-derived macrophages (BMDMs) using Monosodium Urate (MSU), Nigericin, Adenosine Triphosphate (ATP), and Pam3CSK4, separately. Western blot analysis (WB) was employed to detect Pro-caspase-1, Pro-Interleukin-1β (Pro-IL-1β) in cell lysates, and caspase-1, IL-1β in supernatants. Enzyme-Linked Immunosorbent Assay (ELISA) was used to measured the release of IL-1β, IL-18, and Tumor Necrosis Factor-alpha (TNF-α) in cell supernatants to assess the impact of acacetin on NLRP3 inflammasome activation. The lactate dehydrogenase (LDH) release was also assessed. The Nuclear Factor Kappa B (NF-κB) and Mitogen-Activated Protein Kinase (MAPK) signaling pathways related proteins were evaluated by WB, and NF-κB nuclear translocation was observed via laser scanning confocal microscopy (LSCM). Disuccinimidyl Suberate (DSS) cross-linking was employed to detect oligomerization of Apoptosis-associated Speck-like protein containing a Caspase Recruitment Domain (ASC), and LSCM was also used to observe Reactive Oxygen Species (ROS) production. Inductively Coupled Plasma (ICP) and N-(6-methoxyquinolyl) acetoethyl ester (MQAE) assays were utilized to determined the effects of acacetin on the efflux of potassium (K+) and chloride (Cl-) ions. Results: Acacetin inhibited NLRP3 inflammasome activation induced by various agonists, reducing the release of TNF-α, IL-1β, IL-18, and LDH. It suppressed the expression of Lipopolysaccharides (LPS)-activated Phosphorylated ERK (p-ERK), p-JNK, and p-p38, inhibited NF-κB p65 phosphorylation and nuclear translocation. Acacetin also reduced ROS production and inhibited ASC aggregation, thus suppressing NLRP3 inflammasome activation. Notably, acacetin did not affect K+ and Cl-ions efflux during the activation process. Conclusion: Acacetin shows inhibitory effects on both the priming and assembly processes of the NLRP3 inflammasome, positioning it as a promising new candidate for the treatment of NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Juan Bu
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yeledan Mahan
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Shengnan Zhang
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xuanxia Wu
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaoling Zhang
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ling Zhou
- Medical and Translational Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yanmin Zhang
- Scientific Research and Education Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
23
|
Krajewski PK, Tsoukas M, Szepietowski JC. Pathological and Therapeutical Implications of Pyroptosis in Psoriasis and Hidradenitis Suppurativa: A Narrative Review. Curr Issues Mol Biol 2024; 46:663-676. [PMID: 38248345 PMCID: PMC10814322 DOI: 10.3390/cimb46010043] [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: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
This manuscript explores the role of pyroptosis, an inflammatory programmed cell death, in the pathogenesis of two chronic dermatoses, psoriasis and hidradenitis suppurativa (HS). The diseases, though clinically diverse, share common pathogenetic pathways involving the unbalanced interaction between the adaptive and innate immune systems. This review focuses on the molecular changes in psoriatic and HS skin, emphasizing the activation of dendritic cells, secretion of interleukins (IL-17, IL-22, and TNF-α), and the involvement of inflammasomes, particularly NLRP3. This manuscript discusses the role of caspases, especially caspase-1, in driving pyroptosis and highlights the family of gasdermins (GSDMs) as key players in the formation of pores leading to cell rupture and the release of proinflammatory signals. This study delves into the potential therapeutic implications of targeting pyroptosis in psoriasis and HS, examining existing medications like biologics and Janus kinase inhibitors. It also reviews the current limitations and challenges in developing therapies that selectively target pyroptosis. Additionally, the manuscript explores the role of pyroptosis in various inflammatory disorders associated with psoriasis and HS, such as inflammatory bowel disease, diabetes mellitus, and cardiovascular disorders. The review concludes by emphasizing the need for further research to fully elucidate the pathomechanisms of these dermatoses and develop effective, targeted therapies.
Collapse
Affiliation(s)
- Piotr K. Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, 50-368 Wroclaw, Poland;
| | - Maria Tsoukas
- Department of Dermatology, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Jacek C. Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, 50-368 Wroclaw, Poland;
| |
Collapse
|
24
|
Wang L, Zhu Y, Zhang L, Guo L, Wang X, Pan Z, Jiang X, Wu F, He G. Mechanisms of PANoptosis and relevant small-molecule compounds for fighting diseases. Cell Death Dis 2023; 14:851. [PMID: 38129399 PMCID: PMC10739961 DOI: 10.1038/s41419-023-06370-2] [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: 07/04/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Pyroptosis, apoptosis, and necroptosis are mainly programmed cell death (PCD) pathways for host defense and homeostasis. PANoptosis is a newly distinct inflammatory PCD pathway that is uniquely regulated by multifaceted PANoptosome complexes and highlights significant crosstalk and coordination among pyroptosis (P), apoptosis (A), and/or necroptosis(N). Although some studies have focused on the possible role of PANpoptosis in diseases, the pathogenesis of PANoptosis is complex and underestimated. Furthermore, the progress of PANoptosis and related agonists or inhibitors in disorders has not yet been thoroughly discussed. In this perspective, we provide perspectives on PANoptosome and PANoptosis in the context of diverse pathological conditions and human diseases. The treatment targeting on PANoptosis is also summarized. In conclusion, PANoptosis is involved in plenty of disorders including but not limited to microbial infections, cancers, acute lung injury/acute respiratory distress syndrome (ALI/ARDS), ischemia-reperfusion, and organic failure. PANoptosis seems to be a double-edged sword in diverse conditions, as PANoptosis induces a negative impact on treatment and prognosis in disorders like COVID-19 and ALI/ARDS, while PANoptosis provides host protection from HSV1 or Francisella novicida infection, and kills cancer cells and suppresses tumor growth in colorectal cancer, adrenocortical carcinoma, and other cancers. Compounds and endogenous molecules focused on PANoptosis are promising therapeutic strategies, which can act on PANoptosomes-associated members to regulate PANoptosis. More researches on PANoptosis are needed to better understand the pathology of human conditions and develop better treatment.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yanghui Zhu
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Lu Zhang
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Linghong Guo
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xiaoyun Wang
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhaoping Pan
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xian Jiang
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| | - Fengbo Wu
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| | - Gu He
- Department of Dermatology & Venerology and Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
25
|
Vijayapoopathi S, Ramamoorthy R, Meganathan J, Kalaiyazhagan A, Bhuvarahamurthy S, Venugopal B. Nutraceutical combination ameliorates imiquimod-induced psoriasis in mice. Chem Biol Drug Des 2023; 102:1578-1587. [PMID: 37705136 DOI: 10.1111/cbdd.14350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/04/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects both localized and systemic regions of the body. This condition is characterized by the hyperproliferation of keratinocytes, resulting in skin thickening, scaling, and erythema. The severity of psoriasis depends on the extent of skin involvement, the location of the infection, and the symptoms that the person exhibits. While no cure exists, conventional therapies such as topical and systemic drugs are generally used to manage the exacerbation of symptoms. However, chronic use and overdose can lead to other severe adverse effects. Therefore, scientists and researchers are exploring potential nutraceuticals that can be considered as an alternative source of management for psoriasis. Current research aims to use different combinations of natural compounds like cannabidiol, myo-inositol, eicosapentaenoic acid, and krill oil to study the effect of these compounds in the prevention and treatment of psoriasis in the imiquimod (IMQ)-induced psoriatic mice model. The Psoriasis Area Severity Index (PASI) scoring system is used to analyze skin thickness, scales, and erythema. The results indicate that the krill oil combined with the cannabidiol and myo-inositol shows better results than other nutraceutical combinations. In the future, the natural products of krill oil can be combined with cannabidiol and myo-inositol to create an improved alternative to existing steroidal and nonsteroidal anti-inflammatory drugs for psoriasis treatment.
Collapse
Affiliation(s)
- Singaravel Vijayapoopathi
- Department of Medical Biochemistry, Dr. ALM Post-Graduation Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Rajalakshmi Ramamoorthy
- Department of Medical Biochemistry, Dr. ALM Post-Graduation Institute of Basic Medical Sciences, University of Madras, Chennai, India
- Department of Obstetrics, Gynecology and Reproductive Studies, University of Miami, Coral Gables, Florida, USA
| | - Jayaprakash Meganathan
- Department of Medical Biochemistry, Dr. ALM Post-Graduation Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Ananthi Kalaiyazhagan
- Department of Medical Biochemistry, Dr. ALM Post-Graduation Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | | | - Bhuvarahamurthy Venugopal
- Department of Medical Biochemistry, Dr. ALM Post-Graduation Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
26
|
Shaik MG, Joshi SV, Akunuri R, Rana P, Rahman Z, Polomoni A, Yaddanapudi VM, Dandekar MP, Srinivas N. Small molecule inhibitors of NLRP3 inflammasome and GSK-3β in the management of traumatic brain injury: A review. Eur J Med Chem 2023; 259:115718. [PMID: 37573828 DOI: 10.1016/j.ejmech.2023.115718] [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: 11/10/2022] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) is a debilitating mental condition which causes physical disability and morbidity worldwide. TBI may damage the brain by direct injury that subsequently triggers a series of neuroinflammatory events. The activation of NLRP3 inflammasome and dysregulated host immune system has been documented in various neurological disorders such as TBI, ischemic stroke and multiple sclerosis. The activation of NLRP3 post-TBI increases the production of pro-inflammatory cytokines and caspase-1, which are major drivers of neuroinflammation and apoptosis. Similarly, GSK-3β regulates apoptosis through tyrosine kinase and canonical Wnt signalling pathways. Thus, therapeutic targeting of NLRP3 inflammasome and GSK-3β has emerged as promising strategies for regulating the post-TBI neuroinflammation and neurobehavioral disturbances. In this review, we discuss the identification & development of several structurally diverse and pharmacologically interesting small molecule inhibitors for targeting the NLRP3 inflammasome and GSK-3β in the management of TBI.
Collapse
Affiliation(s)
- Mahammad Ghouse Shaik
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ravikumar Akunuri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India; Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Preeti Rana
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India
| | - Anusha Polomoni
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500 037, India.
| | - Nanduri Srinivas
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, India.
| |
Collapse
|
27
|
Son SE, Im DS. Therapeutic effects of candesartan in inflammatory skin disorders by suppressing Th17 differentiation. Int Immunopharmacol 2023; 124:110995. [PMID: 37801970 DOI: 10.1016/j.intimp.2023.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
As angiotensin II is associated with inflammation, type I angiotensin II receptor blockers (ARBs) exibit anti-inflammatory effects in patients with hypertension as well as inflammatory disease animal models including arthritis models. The present study aimed to investigate whether ARBs exert anti-inflammatory effects in vivo in skin disorders. We tested effects of ARBs on 1-chloro-2,4-dinitrobenzene(CDNB)-induced atopic dermatitis-like and imiquimod-induced psoriasis-like skin models. CDNB-induced atopic dermatitis-like skin lesions were suppressed by administration of candesartan or telmisartan. The suppressive effect of telmisartan was blocked by the presence of GW9662, a selective PPARγ inhibitor, but not that of candesartan. Both ARBs suppressed increases in pro-inflammatory cytokine (IL-4, IL-13, IFN-γ, and IL-17A) levels, and GW9662 inhibited telmisartan-induced suppression but not candesartan. Candesartan significantly inhibited in vitro differentiation of naïve T cells into Th17 cells to a greater extent than telmisartan. In the imiquimod-induced psoriasis model, whose primary etiology is activation of IL-23/IL-17 axis, candesartan significantly suppressed psoriasis-like skin lesions and Th17 cell populations in both lymph nodes and spleens to a greater extent than telmisartan. Overall, certain ARBs may have anti-inflammatory effects in skin diseases. Candesartan may have therapeutic implications in inflammatory skin disorders by suppressing Th17 differentiation, while telmisartan might have therapeutic potential by activating PPARγ.
Collapse
Affiliation(s)
- So-Eun Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
28
|
Zheng XQ, Kong XQ, He Y, Wang YJ, Xie L, Liu LL, Lin LR, Yang TC. Treponema pallidum recombinant protein Tp47 enhanced interleukin-6 secretion in human dermal fibroblasts through the toll-like receptor 2 via the p38, PI3K/Akt, and NF-κB signalling pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119540. [PMID: 37468070 DOI: 10.1016/j.bbamcr.2023.119540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Interleukin-6 (IL-6) is a multi-effective cytokine involved in multiple immune responses. Whether fibroblasts also turn out to be a cytokine IL-6 factory during interaction with Treponema pallidum is not yet understood. To explore whether fibroblasts participate in inflammation due to syphilis, a series of experiments were performed to explore the role of T. pallidum lipoprotein Tp47 in IL-6 production in human dermal fibroblasts. The Toll-like receptor 2 (TLR2) and participating signalling pathways in this process were also evaluated. The results showed that the expressions of IL-6 and the protein levels of TLR2 in fibroblasts were upregulated after stimulation with Tp47, and this effect was impeded by the TLR2 inhibitor C29. In addition, Tp47 promoted the phosphorylation of p38, PI3K/Akt, and nuclear factor-kappaB (NF-κB), and the translocation of NF-κB in fibroblasts. Moreover, p38, PI3K, and NF-κB inhibitors significantly reduced IL-6 production in fibroblasts stimulated with Tp47. Furthermore, the TLR2 inhibitor C29 inhibited the phosphorylation of p38, Akt, and NF-κB, and the translocation of NF-κB in fibroblasts. In conclusion, our results showed that Tp47 enhanced IL-6 secretion in human dermal fibroblasts through TLR2 via p38, PI3K/Akt, and NF-κB signalling pathways. These findings contribute to our understanding of syphilis inflammation.
Collapse
Affiliation(s)
- Xin-Qi Zheng
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xiang-Qi Kong
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Department of Clinical Laboratory, Weifang People's Hospital, Weifang, Shandong 261000, China
| | - Yun He
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Department of Medical Laboratory, The Second Affiliated Hospital of Xiamen Medical College, Xiamen 361004, China
| | - Yong-Jing Wang
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Lin Xie
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Li-Li Liu
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen 361004, China.
| | - Li-Rong Lin
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen 361004, China.
| | - Tian-Ci Yang
- Center of Clinical Laboratory, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen 361004, China.
| |
Collapse
|
29
|
Dai Y, Zhou J, Shi C. Inflammasome: structure, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e391. [PMID: 37817895 PMCID: PMC10560975 DOI: 10.1002/mco2.391] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammasomes are a group of protein complex located in cytoplasm and assemble in response to a wide variety of pathogen-associated molecule patterns, damage-associated molecule patterns, and cellular stress. Generally, the activation of inflammasomes will lead to maturation of proinflammatory cytokines and pyroptotic cell death, both associated with inflammatory cascade amplification. A sensor protein, an adaptor, and a procaspase protein interact through their functional domains and compose one subunit of inflammasome complex. Under physiological conditions, inflammasome functions against pathogen infection and endogenous dangers including mtROS, mtDNA, and so on, while dysregulation of its activation can lead to unwanted results. In recent years, advances have been made to clarify the mechanisms of inflammasome activation, the structural details of them and their functions (negative/positive) in multiple disease models in both animal models and human. The wide range of the stimuli makes the function of inflammasome diverse and complex. Here, we review the structure, biological functions, and therapeutic targets of inflammasomes, while highlight NLRP3, NLRC4, and AIM2 inflammasomes, which are the most well studied. In conclusion, this review focuses on the activation process, biological functions, and structure of the most well-studied inflammasomes, summarizing and predicting approaches for disease treatment and prevention with inflammasome as a target. We aim to provide fresh insight into new solutions to the challenges in this field.
Collapse
Affiliation(s)
- Yali Dai
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| | - Jing Zhou
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
- Institute of ImmunologyArmy Medical UniversityChongqingChina
| | - Chunmeng Shi
- Institute of Rocket Force MedicineState Key Laboratory of Trauma and Chemical PoisoningArmy Medical UniversityChongqingChina
| |
Collapse
|
30
|
Spangenberg SH, Palermo A, Gazaniga NR, Martínez-Peña F, Guijas C, Chin EN, Rinschen MM, Sander PN, Webb B, Pereira LE, Jia Y, Meitz L, Siuzdak G, Lairson LL. Hydroxyproline metabolism enhances IFN-γ-induced PD-L1 expression and inhibits autophagic flux. Cell Chem Biol 2023; 30:1115-1134.e10. [PMID: 37467751 PMCID: PMC11426993 DOI: 10.1016/j.chembiol.2023.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/20/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
The immune checkpoint protein PD-L1 plays critical roles in both immune system homeostasis and tumor progression. Impaired PD-1/PD-L1 function promotes autoimmunity and PD-L1 expression within tumors promotes immune evasion. If and how changes in metabolism or defined metabolites regulate PD-L1 expression is not fully understood. Here, using a metabolomics activity screening-based approach, we have determined that hydroxyproline (Hyp) significantly and directly enhances adaptive (i.e., IFN-γ-induced) PD-L1 expression in multiple relevant myeloid and cancer cell types. Mechanistic studies reveal that Hyp acts as an inhibitor of autophagic flux, which allows it to regulate this negative feedback mechanism, thereby contributing to its overall effect on PD-L1 expression. Due to its prevalence in fibrotic tumors, these findings suggest that hydroxyproline could contribute to the establishment of an immunosuppressive tumor microenvironment and that Hyp metabolism could be targeted to pharmacologically control PD-L1 expression for the treatment of cancer or autoimmune diseases.
Collapse
Affiliation(s)
| | - Amelia Palermo
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nathalia R Gazaniga
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Emily N Chin
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Markus M Rinschen
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Philipp N Sander
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bill Webb
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Laura E Pereira
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ying Jia
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lance Meitz
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, the Scripps Research Institute, La Jolla, CA 92037, USA; Department of Integrative Structural and Computational Biology, La Jolla, CA 92037, USA.
| | - Luke L Lairson
- Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
31
|
Shahi A, Afzali S, Amirzargar A, Mohaghegh P, Salehi S, Mansoori Y. Potential roles of inflammasomes in the pathophysiology of Psoriasis: A comprehensive review. Mol Immunol 2023; 161:44-60. [PMID: 37481828 DOI: 10.1016/j.molimm.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/20/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023]
Abstract
Psoriasis is an inflammatory skin disease whose pathophysiology is attributed to both innate and adaptive immune cells and molecules. Despite the crucial roles of the immune system in psoriasis, it cannot be categorized as an autoimmune disease because of the lack of main signs of autoimmunity, such as specific antibodies, well-defined antigens, and autoimmune genetic risk factors. The presence of some cellular and molecular properties, such as the presence of neutrophils in skin lesions and the activation of the innate immune system, attributes psoriasis to a group of diseases called autoinflammatory disorders. Autoinflammatory diseases refer to a group of inherited disorders whose main manifestations are recurrent fever, a high level of acute-phase reactant, and a tendency for inflammation of the skin, joints, and other organs like the nervous system. In most autoinflammatory disorders, it has been seen that complexes of the high-molecular-weight protein named inflammasomes have significant roles. The inflammasome complex usually is formed and activated in the stimulated immune cell cytoplasm, and its activation consequently leads to inflammatory events such as producing of active caspase-1, mature interleukin-1β (IL-1β), and IL-18 and can cause an inflammatory programmed cell death called pyroptosis. Since the identification of inflammasomes, it has been shown that there are close links between them and hereditary and acquired autoinflammatory diseases like psoriasis. In this review, we aim to focus on well-defined inflammasome and their role in the pathophysiology of psoriasis.
Collapse
Affiliation(s)
- Abbas Shahi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Afzali
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Poopak Mohaghegh
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Saeedeh Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
32
|
Ajetunmobi OH, Wall G, Vidal Bonifacio B, Martinez Delgado LA, Chaturvedi AK, Najvar LK, Wormley FL, Patterson HP, Wiederhold NP, Patterson TF, Lopez-Ribot JL. High-Throughput Screening of the Repurposing Hub Library to Identify Drugs with Novel Inhibitory Activity against Candida albicans and Candida auris Biofilms. J Fungi (Basel) 2023; 9:879. [PMID: 37754987 PMCID: PMC10532723 DOI: 10.3390/jof9090879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
Candidiasis is one of the most frequent nosocomial infections affecting an increasing number of at-risk patients. Candida albicans remains the most frequent causative agent of candidiasis, but, in the last decade, C. auris has emerged as a formidable multi-drug-resistant pathogen. Both species are fully capable of forming biofilms, which contribute to resistance, increasing the urgency for new effective antifungal therapies. Repurposing existing drugs could significantly accelerate the development of novel therapies against candidiasis. Here, we have screened the Repurposing Hub library from the Broad Institute, containing over 6000 compounds, in search for inhibitors of C. albicans and C. auris biofilm formation. The primary screen identified 57 initial hits against C. albicans and 33 against C. auris. Confirmatory concentration-dependent assays were used to validate the activity of the initial hits and, at the same time, establish their anti-biofilm potency. Based on these results, ebselen, temsirolimus, and compound BAY 11-7082 emerged as the leading repositionable compounds. Subsequent experiments established their spectrum of antifungal activity against yeasts and filamentous fungi. In addition, their in vivo activity was examined in the murine models of hematogenously disseminated C. albicans and C. auris infections. Although promising, further in vitro and in vivo studies are needed to confirm their potential use for the therapy of candidiasis and possibly other fungal infections.
Collapse
Affiliation(s)
- Olabayo H. Ajetunmobi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Gina Wall
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Bruna Vidal Bonifacio
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | | | - Ashok K. Chaturvedi
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| | - Laura K. Najvar
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (L.K.N.); (T.F.P.)
| | - Floyd L. Wormley
- Department of Biology, Texas Christian University, Fort Worth, TX 76129, USA;
| | - Hoja P. Patterson
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (H.P.P.); (N.P.W.)
| | - Nathan P. Wiederhold
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (H.P.P.); (N.P.W.)
| | - Thomas F. Patterson
- Department of Medicine, Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (L.K.N.); (T.F.P.)
| | - Jose L. Lopez-Ribot
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (O.H.A.); (A.K.C.)
| |
Collapse
|
33
|
Liu Y, Lei H, Zhang W, Xing Q, Liu R, Wu S, Liu Z, Yan Q, Li W, Liu X, Hu Y. Pyroptosis in renal inflammation and fibrosis: current knowledge and clinical significance. Cell Death Dis 2023; 14:472. [PMID: 37500614 PMCID: PMC10374588 DOI: 10.1038/s41419-023-06005-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Pyroptosis is a novel inflammatory form of regulated cell death (RCD), characterized by cell swelling, membrane rupture, and pro-inflammatory effects. It is recognized as a potent inflammatory response required for maintaining organismal homeostasis. However, excessive and persistent pyroptosis contributes to severe inflammatory responses and accelerates the progression of numerous inflammation-related disorders. In pyroptosis, activated inflammasomes cleave gasdermins (GSDMs) and generate membrane holes, releasing interleukin (IL)-1β/18, ultimately causing pyroptotic cell death. Mechanistically, pyroptosis is categorized into caspase-1-mediated classical pyroptotic pathway and caspase-4/5/11-mediated non-classical pyroptotic pathway. Renal fibrosis is a kidney disease characterized by the loss of structural and functional units, the proliferation of fibroblasts and myofibroblasts, and extracellular matrix (ECM) accumulation, which leads to interstitial fibrosis of the kidney tubules. Histologically, renal fibrosis is the terminal stage of chronic inflammatory kidney disease. Although there is a multitude of newly discovered information regarding pyroptosis, the regulatory roles of pyroptosis involved in renal fibrosis still need to be fully comprehended, and how to improve clinical outcomes remains obscure. Hence, this review systematically summarizes the novel findings regarding the role of pyroptosis in the pathogenesis of renal fibrosis and discusses potential biomarkers and drugs for anti-fibrotic therapeutic strategies.
Collapse
Affiliation(s)
- Ya Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Haibo Lei
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wenyou Zhang
- Department of Pharmacy, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qichang Xing
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Renzhu Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Shiwei Wu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Zheng Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qingzi Yan
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wencan Li
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Xiang Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| | - Yixiang Hu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| |
Collapse
|
34
|
Chen J, Li Q, Hong Y, Zhou X, Yu C, Tian X, Zhao J, Long C, Shen L, Wu S, Wei G. Inhibition of the NF-κB Signaling Pathway Alleviates Pyroptosis in Bladder Epithelial Cells and Neurogenic Bladder Fibrosis. Int J Mol Sci 2023; 24:11160. [PMID: 37446339 DOI: 10.3390/ijms241311160] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/25/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Most children with a neurogenic bladder (NB) have bladder fibrosis, which causes irreversible bladder dysfunction and damage to the upper urinary tract. However, the mechanism of bladder fibrosis remains unclear. This study aimed to investigate the underlying causes of bladder fibrosis. Here, the lumbar 6 (L6) and sacral 1 (S1) spinal nerves of Sprague Dawley rats were severed bilaterally to establish NB models. Using RNA-seq, we discovered that the NF-κB signaling pathway and inflammation were upregulated in spinal cord injury (SCI)-induced bladder fibrosis. Subsequent Western blotting, enzyme-linked immunosorbent assays, immunohistochemical staining, and immunofluorescence staining verified the RNA-seq findings. To further clarify whether the NF-κB signaling pathway and pyroptosis were involved in bladder fibrosis, a TGF-β1-treated urinary epithelial cell line (SV-HUC-1 cells) was used as an in vitro model. Based on the results of RNA-seq, we consistently found that the NF-κB signaling pathway and pyroptosis might play important roles in TGF-β1-treated cells. Further experiments also confirmed the RNA-seq findings in vitro. Moreover, using the NLRP3 inhibitor MCC950 rescued TGF-β1-induced fibrosis, and the NF-κB signaling pathway inhibitor BAY 11-7082 effectively rescued TGF-β1-induced pyroptosis and the deposition of extracellular matrix by SV-HUC-1 cells. In summary, our research demonstrated for the first time that the NF-κB signaling pathway inhibition rescued bladder epithelial cells pyroptosis and fibrosis in neurogenic bladders.
Collapse
Affiliation(s)
- Jing Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Qi Li
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Yifan Hong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Xiazhu Zhou
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Xiaomao Tian
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Jie Zhao
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Chunlan Long
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Lianju Shen
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, China
- Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| |
Collapse
|
35
|
Vaher H, Kingo K, Kolberg P, Pook M, Raam L, Laanesoo A, Remm A, Tenson T, Alasoo K, Mrowietz U, Weidinger S, Kingo K, Rebane A. Skin Colonization with S. aureus Can Lead to Increased NLRP1 Inflammasome Activation in Patients with Atopic Dermatitis. J Invest Dermatol 2023; 143:1268-1278.e8. [PMID: 36736455 DOI: 10.1016/j.jid.2023.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/16/2022] [Accepted: 01/04/2023] [Indexed: 02/04/2023]
Abstract
The role of NLRP1 inflammasome activation and subsequent production of IL-1 family cytokines in the development of atopic dermatitis (AD) is not clearly understood. Staphylococcus aureus is known to be associated with increased mRNA levels of IL1 family cytokines in the skin and more severe AD. In this study, the altered expression of IL-1 family cytokines and inflammasome-related genes was confirmed, and a positive relationship between mRNA levels of inflammasome sensor NLRP1 and IL1B or IL18 was determined. Enhanced expression of the NLRP1 and PYCARD proteins and increased caspase-1 activity were detected in the skin of patients with AD. The genetic association of IL18R1 and IL18RAP with AD was confirmed, and the involvement of various immune cell types was predicted using published GWAS and expression quantitative trait loci datasets. In keratinocytes, the inoculation with S. aureus led to the increased secretion of IL-1β and IL-18, whereas small interfering RNA silencing of NLRP1 inhibited the production of these cytokines. Our results suggest that skin colonization with S. aureus may cause the activation of the NLRP1 inflammasome in keratinocytes, which leads to the secretion of IL-1β and IL-18 and thereby may contribute to the pathogenesis of AD, particularly in the presence of genetic variations in the IL-18 pathway.
Collapse
Affiliation(s)
- Helen Vaher
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kristiina Kingo
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Peep Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Martin Pook
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Liisi Raam
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia; Department of Dermatology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Anet Laanesoo
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Anu Remm
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tanel Tenson
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Ulrich Mrowietz
- Department of Dermatology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Stephan Weidinger
- Department of Dermatology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Külli Kingo
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia; Department of Dermatology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| |
Collapse
|
36
|
Wang H, Jin H, Liu Z, Tan C, Wei L, Fu M, Huang Y. Screening and identification of key chromatin regulator biomarkers for ankylosing spondylitis and drug prediction: evidence from bioinformatics analysis. BMC Musculoskelet Disord 2023; 24:389. [PMID: 37193965 DOI: 10.1186/s12891-023-06490-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Ankylosing spondylitis (AS) is one of the most common immune-mediated arthritic diseases worldwide. Despite considerable efforts to elucidate its pathogenesis, the molecular mechanisms underlying AS are still not fully understood. METHODS To identify candidate genes involved in AS progression, the researchers downloaded the microarray dataset GSE25101 from the Gene Expression Omnibus (GEO) database. They identified differentially expressed genes (DEGs) and functionally enriched them for analysis. They also constructed a protein-protein interaction network (PPI) using STRING and performed cytoHubba modular analysis, immune cell and immune function analysis, functional analysis and drug prediction.The results showed that DEGs were mainly associated with histone modifications, chromatin organisation, transcriptional coregulator activity, transcriptional co-activator activity, histone acetyltransferase complexes and protein acetyltransferase complexes. RESULTS The researchers analysed the differences in expression between the CONTROL and TREAT groups in terms of immunity to determine their effect on TNF-α secretion. By obtaining hub genes, they predicted two therapeutic agents, AY 11-7082 and myricetin. CONCLUSION The DEGs, hub genes and predicted drugs identified in this study contribute to our understanding of the molecular mechanisms underlying the onset and progression of AS. They also provide candidate targets for the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Han Wang
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Hongbo Jin
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Zhiyang Liu
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Chengju Tan
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Lin Wei
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Mingfen Fu
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China
| | - Yizhuan Huang
- Acupuncture and Massage Department, Affiliated Sport Hospital of CDSU, number 251, Wu Hou Ci Da Jie, Cheng Du, Si Chuan, 610041, China.
| |
Collapse
|
37
|
Gu Q, Zou J, Zhou Y, Deng Q. Mechanism of inflammasomes in cancer and targeted therapies. Front Oncol 2023; 13:1133013. [PMID: 37020871 PMCID: PMC10067570 DOI: 10.3389/fonc.2023.1133013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Inflammasomes, composed of the nucleotide-binding oligomerization domain(NOD)-like receptors (NLRs), are immune-functional protein multimers that are closely linked to the host defense mechanism. When NLRs sense pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), they assemble into inflammasomes. Inflammasomes can activate various inflammatory signaling pathways, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, and produce a large number of proinflammatory cytokines, which are closely associated with multiple cancers. They can also accelerate the occurrence and development of cancer by providing suitable tumor microenvironments, promoting tumor cell proliferation, and inhibiting tumor cell apoptosis. Therefore, the exploitation of novel targeted drugs against various inflammasomes and proinflammatory cytokines is a new idea for the treatment of cancer. In recent years, more than 50 natural extracts and synthetic small molecule targeted drugs have been reported to be in the research stage or have been applied to the clinic. Herein, we will overview the mechanisms of inflammasomes in common cancers and discuss the therapeutic prospects of natural extracts and synthetic targeted agents.
Collapse
Affiliation(s)
- Qingdan Gu
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Jiazhen Zou
- Department of Laboratory Medicine, Shenzhen Second People’s Hospital, The First Affiliated 5 Hospital of Shenzhen University, Health Science Center, Shenzhen, China
| | - Ying Zhou
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Qiuchan Deng
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
- *Correspondence: Qiuchan Deng,
| |
Collapse
|
38
|
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol 2023; 16:24. [PMID: 36932407 PMCID: PMC10022228 DOI: 10.1186/s13045-023-01407-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Inflammasomes are macromolecular platforms formed in response to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns, whose formation would cause maturation of interleukin-1 (IL-1) family members and gasdermin D (GSDMD), leading to IL-1 secretion and pyroptosis respectively. Several kinds of inflammasomes detecting different types of dangers have been found. The activation of inflammasomes is regulated at both transcription and posttranscription levels, which is crucial in protecting the host from infections and sterile insults. Present findings have illustrated that inflammasomes are involved in not only infection but also the pathology of tumors implying an important link between inflammation and tumor development. Generally, inflammasomes participate in tumorigenesis, cell death, metastasis, immune evasion, chemotherapy, target therapy, and radiotherapy. Inflammasome components are upregulated in some tumors, and inflammasomes can be activated in cancer cells and other stromal cells by DAMPs, chemotherapy agents, and radiation. In some cases, inflammasomes inhibit tumor progression by initiating GSDMD-mediated pyroptosis in cancer cells and stimulating IL-1 signal-mediated anti-tumor immunity. However, IL-1 signal recruits immunosuppressive cell subsets in other cases. We discuss the conflicting results and propose some possible explanations. Additionally, we also summarize interventions targeting inflammasome pathways in both preclinical and clinical stages. Interventions targeting inflammasomes are promising for immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Ziqi Zhang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
39
|
Liu CT, Yen JHJ, Brown DA, Song YC, Chu MY, Hung YH, Tang YH, Wu PY, Yen HR. Targeting Nrf2 with 3 H-1,2-dithiole-3-thione to moderate OXPHOS-driven oxidative stress attenuates IL-17A-induced psoriasis. Biomed Pharmacother 2023; 159:114294. [PMID: 36706632 DOI: 10.1016/j.biopha.2023.114294] [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: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Psoriasis, a chronic autoimmune disease characterized by the hyperproliferation of keratinocytes in the epidermis and parakeratosis, significantly impacts quality of life. Interleukin (IL)- 17A dominates the pathogenesis of psoriasis and facilitates reactive oxygen species (ROS) accumulation, which exacerbates local psoriatic lesions. Biologic treatment provides remarkable clinical efficacy, but its high cost and unignorable side effects limit its applications. 3 H-1,2-Dithiole-3-thione (D3T) possesses compelling antioxidative capacities against several diseases through the nuclear factor erythroid 2-related factor 2 (Nrf2) cascade. Hence, we aimed to evaluate the effect and mechanism of D3T in psoriasis. We found that D3T attenuates skin thickening and scaling by inhibiting IL-17A-secreting γδT cells in imiquimod (IMQ)-induced psoriatic mice. Interleukin-17A markedly enhanced IL-6 and IL-8 expression, lipid peroxidation, the contents of nitric oxide and hydrogen peroxide, oxidative phosphorylation and the MAPK/NF-κB pathways in keratinocytes. IL-17A also inhibited the Nrf2-NQO1-HO-1 axis and the activities of superoxide dismutase and glutathione peroxidase. D3T significantly reversed these parameters in IL-17A-treated keratinocytes. ML-385, a Nrf2 neutralizer, failed to improve D3T-induced anti-inflammatory and antioxidative effects in IL-17A-treated keratinocytes. We conclude that targeting Nrf2 with D3T to diminish oxidative and inflammatory damage in keratinocytes may attenuate psoriasis.
Collapse
Affiliation(s)
- Chuan-Teng Liu
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jui-Hung Jimmy Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, USA
| | - Dennis A Brown
- Manchester University College of Pharmacy, Natural and Health Sciences, Fort Wayne, IN, USA
| | - Ying-Chyi Song
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Mei-Yun Chu
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Yu-Hsiang Hung
- Department of Medical Education, China Medical University Hospital, Taichung, Taiwan
| | | | - Po-Yuan Wu
- Department of Dermatology, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| | - Hung-Rong Yen
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Medical Biotechnology and Laboratory Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
40
|
Chen X, Deng Q, Li X, Xian L, Xian D, Zhong J. Natural Plant Extract - Loganin: A Hypothesis for Psoriasis Treatment Through Inhibiting Oxidative Stress and Equilibrating Immunity via Regulation of Macrophage Polarization. Clin Cosmet Investig Dermatol 2023; 16:407-417. [PMID: 36817639 PMCID: PMC9936880 DOI: 10.2147/ccid.s396173] [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: 11/04/2022] [Accepted: 01/12/2023] [Indexed: 02/16/2023]
Abstract
Psoriasis, a chronic immune-mediated inflammatory skin disease, influences approximately 2-3% of the world's population. At present, the etiology of psoriasis remains unclear and there is still no causal treatment available. Recent studies indicate that oxidative stress (OS) and T cells dysregulation may participate in the pathogenesis of psoriasis, among which M1-dominant macrophage polarization is a crucial contributor. Macrophages mainly polarize into two different subsets, ie, classically activated macrophage (M1) and alternatively activated macrophage (M2). M1 polarization tends to exacerbate psoriasis via producing substantial reactive oxygen species (ROS) and inflammatory mediators, to encourage OS invasion and T cells dysregulation. Thus, targeting M1 polarization can be a possible therapeutic alternative for psoriasis. Loganin, belonging to iridoid glycosides, is a pharmaceutically active ingredient originated from Cornus officinalis, exerting multiple biological activities, eg, immunomodulation, antioxidation, anti-inflammation, etc. More importantly, it could effectively suppress M1 polarization, thereby arresting OS aggression and T cells' dysregulation. Numerous studies have confirmed that loganin is quite reliable for diseases treatment via suppressing M1 polarization. Nevertheless, reports about loganin treating psoriasis have seldom appeared so far. Accordingly, we hold a hypothesis that loganin would availably manage psoriasis through preventing M1 polarization. Data from previous studies guarantee the potential of loganin in control of psoriasis.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Qiyan Deng
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xiaolong Li
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Li Xian
- Department of Emergency, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Dehai Xian
- Department of Anatomy, Southwest Medical University, Luzhou, 646000, People’s Republic of China,Correspondence: Jianqiao Zhong, Email ; Dehai Xian, Email
| | - Jianqiao Zhong
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China,Correspondence: Jianqiao Zhong, Email ; Dehai Xian, Email
| |
Collapse
|
41
|
Wu X, Yin Q, Wang J, Dai C, Wang J, Guo X, Jiang F. Novel RNA polymerase I inhibitor CX-5461 suppresses imiquimod-induced experimental psoriasis. Exp Dermatol 2023; 32:91-99. [PMID: 36168732 DOI: 10.1111/exd.14682] [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: 05/28/2022] [Revised: 08/31/2022] [Accepted: 09/25/2022] [Indexed: 01/06/2023]
Abstract
Clinical treatment of psoriasis remains challenging because of possible long-term drug toxicities and loss of therapeutic effects over time. CX-5461 is a novel selective inhibitor of RNA polymerase I. Our previous studies have shown that CX-5461 has potent anti-inflammatory effects. Here we investigated whether CX-5461 could inhibit the development of imiquimod-induced experimental psoriasis in mice. Adult male C57BL/6 mice were used, and psoriasis-like lesions were induced by topical imiquimod treatment. In vivo, we demonstrated that topical application of CX-5461 prevented the development of imiquimod-induced psoriasis, with decreases in keratinocyte proliferation, T-cell infiltration and pathological angiogenesis. CX-5461 also reversed existing skin inflammation induced imiquimod and retarded the development of 12-O-tetradecanoylphorbol-13-acetate-induced epidermal hyperplasia and inflammation. In vitro, CX-5461 induced cell cycle arrest in keratinocytes, inhibited expressions of interleukin-17, interleukin-23 receptor and retinoic acid receptor-related orphan receptor-γt in activated T cells, and reduced angiogenic functions of endothelial cells. In conclusion, CX-5461 exhibits therapeutic effects on experimental psoriasis in mice, likely via multiple mechanisms including anti-proliferative, anti-inflammatory and anti-angiogenic activities.
Collapse
Affiliation(s)
- Xiao Wu
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qihui Yin
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Wang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaochao Dai
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianli Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Jiang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
42
|
Kan W, Li Q, Li P, Ren L, Mu W, Lin L, Wen J, Ge F, Hou M, Hui S, He P, Liang L, Xu Y, Li X, Xu G, Xiao X, Bai Z. Glycyrrhiza uralensis polysaccharides ameliorate acute lung injury by inhibiting the activation of multiple inflammasomes. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
43
|
Chen X, Chen Y, Ou Y, Min W, Liang S, Hua L, Zhou Y, Zhang C, Chen P, Yang Z, Hu W, Sun P. Bortezomib inhibits NLRP3 inflammasome activation and NF-κB pathway to reduce psoriatic inflammation. Biochem Pharmacol 2022; 206:115326. [DOI: 10.1016/j.bcp.2022.115326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/02/2022]
|
44
|
Yang Q, Zhang J, Liu F, Chen H, Zhang W, Yang H, He N, Dong J, Zhao P. A. caviae infection triggers IL-1β secretion through activating NLRP3 inflammasome mediated by NF-κB signaling pathway partly in a TLR2 dependent manner. Virulence 2022; 13:1486-1501. [PMID: 36040120 PMCID: PMC9450903 DOI: 10.1080/21505594.2022.2116169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aeromonas caviae, an important food-borne pathogen, induces serious invasive infections and inflammation. The pro-inflammatory IL-1β functions against pathogenic infections and is elevated in various Aeromonas infection cases. However, the molecular mechanism of A. caviae-mediated IL-1β secretion remains unknown. In this study, mouse macrophages (PMs) were used to establish A. caviae infection model and multiple strategies were utilized to explore the mechanism of IL-1β secretion. IL-1β was elevated in A. caviae infected murine serum, PMs lysates or supernatants. This process triggered NLRP3 levels upregulation, ASC oligomerization, as well as dot gathering of NLRP3 and speck-like signals of ASC in the cytoplasm. MCC950 blocked A. caviae mediated IL-1β release. Meanwhile, NLRP3 inflammasome mediated the release of IL-1β in dose- and time-dependent manners, and the release of IL-1β was dependent on active caspase-1, as well as NLRP3 inflammasome was activated by potassium efflux and cathepsin B release ways. A. caviae also enhanced TLR2 levels, and deletion of TLR2 obviously decreased IL-1β secretion. What’s more, A. caviae resulted in NF-κB p65 nuclear translocation partly in a TLR2-dependent manner. Blocking NF-κB using BAY 11-7082 almost completely inhibited NLRP3 inflammasome first signal pro-IL-1β expression. Blocking TLR2, NF-κB, NLRP3 inflammasome significantly downregulated IL-1β release and TNF-α and IL-6 levels. These data illustrate that A. caviae caused IL-1β secretion in PMs is controlled by NLRP3 inflammasome, of which is mediated by NF-κB pathway and is partially dependent on TLR2, providing basis for drugs against A. caviae.
Collapse
Affiliation(s)
- Qiankun Yang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China.,Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Jianguo Zhang
- Department of Radiation, The Second People's Hospital of Lianyungang (Lianyungang Tumor Hospital), Lianyungang, Jiangsu 222000, China
| | - Feixue Liu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China.,Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Huizhen Chen
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Haitao Yang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Nana He
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Panpan Zhao
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
45
|
Sun L, Yu G, Jiang H, Shi K, Huang D, Bao H, Huang Y, Che L, Chen M. Experimental study on the anti-inflammatory effect of Mongolian medicine Sendeng Decoction on mice with psoriasis. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
46
|
Mi Y, Liang L, Xu K, Li Q, Wang W, Dang W, Deng J, Zhi Y, Li X, Tan J. Severe acute respiratory syndrome coronavirus 2 virus-like particles induce dendritic cell maturation and modulate T cell immunity. Front Cell Infect Microbiol 2022; 12:986350. [DOI: 10.3389/fcimb.2022.986350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that play an important role in both innate and acquired immune responses against pathogens. However, the role of DCs in coronavirus disease 2019 (COVID-19) is unclear. Virus-like particles (VLPs) that structurally mimic the original virus are one of the candidates COVID-19 vaccines. In the present study, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VLPs were used as an alternative to live virus to evaluate the interaction of the virus with DCs. The results revealed that SARS-CoV-2 VLPs induced DC maturation by augmenting cell surface molecule expression (CD80, CD86, and major histocompatibility complex class II (MHC-II)) and inflammatory cytokine production (tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-12p70) in DCs via the mitogen-activated protein kinase and nuclear factor-κB signaling pathways. In addition, mature DCs induced by SARS-CoV-2 VLPs promoted T cell proliferation, which was dependent on VLPs concentration. Our results suggest that SARS-CoV-2 VLPs regulate the immune response by interacting with DCs. These findings will improve the understanding of SARS-CoV-2 pathogenesis and SARS-CoV-2 vaccine development.
Collapse
|
47
|
Proteins from Modern and Ancient Wheat Cultivars: Impact on Immune Cells of Healthy Individuals and Patients with NCGS. Nutrients 2022; 14:nu14204257. [PMID: 36296938 PMCID: PMC9611902 DOI: 10.3390/nu14204257] [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: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
In non-celiac gluten sensitivity (NCGS), the elimination of wheat results in a clear symptom improvement, but gluten has still not been proven as (the sole) trigger. Due to the increase in the prevalence of gluten-related diseases, the breeding of high-performance wheat cultivars is discussed as a trigger. To analyze the immune stimulation and signal pathways, the immune cells of healthy subjects and patients with NCGS were stimulated with gliadins from wheat, and the expression and secretion of interleukin 1ß (IL1ß) and interleukin 6 (IL6) were studied. To determine the impact of wheat breeding, the monocyte cell line THP1 and human immune cells were stimulated with gliadin, glutenin, and albumin/globulin fractions of ancient and modern cereals, and expression of inflammatory molecules was checked. Immune cells of patients with NCGS showed an increased expression of IL1ß and IL6 after stimulation with gliadins compared to immune cells of healthy controls. Gliadins caused a strong activation of P-STAT3 in immune cells of healthy controls, and inhibitors of JAK and NFκB pathways considerably reduced this response. In addition to gliadins, we further showed that glutenins and albumin/globulins from all wheat cultivars from the last century, and especially from einkorn and spelt, also markedly induced the expression of inflammatory genes in THP1 and human immune cells. There was no correlation between enhanced immune stimulation and ancient or modern cultivars. This does not support the hypothesis that modern wheat breeding is responsible for the increase in gluten-related diseases. An altered immune situation is suggested in patients with NCGS.
Collapse
|
48
|
Pizzimenti C, Fiorentino V, Ieni A, Martini M, Tuccari G, Lentini M, Fadda G. Aggressive variants of follicular cell-derived thyroid carcinoma: an overview. Endocrine 2022; 78:1-12. [PMID: 35864338 DOI: 10.1007/s12020-022-03146-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/12/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE The incidence of thyroid carcinoma has increased globally in the past years. Papillary thyroid carcinoma (PTC) is the most frequent neoplasm of the thyroid gland comprehending the 90% of the thyroid carcinoma and has an indolent clinical behaviour. However, some variants of follicular cell-derived thyroid carcinoma, including variants of classic of PTC, have been identified that show a more aggressive biological behaviour. An accurate diagnosis of these entities is crucial for planning a more aggressive treatment and improving patients' prognosis of patients. The aim of this review is to present the main clinical, histological, and molecular features of aggressive variants of follicular cell-derived thyroid carcinoma, and to provide useful histological parameters for determining the most suitable therapeutic strategy for patients affected by these forms. RESULTS Variants of classic PTC such as the diffuse sclerosing variant (DSV), the tall cell variant (TCV), the columnar cell variant (CCV), the solid/trabecular variant (STV) and the hobnail variant (HV), and other variants of follicular cell-derived thyroid carcinoma, such as poorly differentiated thyroid carcinoma (PDTC), and anaplastic thyroid carcinoma (ATC), are associated with aggressive behaviour. CONCLUSIONS The correct identification and diagnosis of aggressive variants of follicular cell-derived thyroid carcinoma is important, as they allow the clinician to adopt the most refined therapeutic strategies in order to the survival of the patients.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98124, Messina, Italy.
| | - Vincenzo Fiorentino
- Department of Pathology, Foundation "Agostino Gemelli", University Hospital IRCCS, 00168, Rome, Italy
| | - Antonio Ieni
- Department of Human Pathology of the Adulthood and Developing Age "Gaetano Barresi", Section of Pathology, University of Messina, 98124, Messina, Italy
| | - Maurizio Martini
- Department of Human Pathology of the Adulthood and Developing Age "Gaetano Barresi", Section of Pathology, University of Messina, 98124, Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology of the Adulthood and Developing Age "Gaetano Barresi", Section of Pathology, University of Messina, 98124, Messina, Italy
| | - Maria Lentini
- Department of Human Pathology of the Adulthood and Developing Age "Gaetano Barresi", Section of Pathology, University of Messina, 98124, Messina, Italy
| | - Guido Fadda
- Department of Human Pathology of the Adulthood and Developing Age "Gaetano Barresi", Section of Pathology, University of Messina, 98124, Messina, Italy
| |
Collapse
|
49
|
Jewell S, Herath AM, Gordon R. Inflammasome Activation in Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S113-S128. [PMID: 35848038 PMCID: PMC9535572 DOI: 10.3233/jpd-223338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Chronic sterile inflammation and persistent immune activation is a prominent pathological feature of Parkinson’s disease (PD). Inflammasomes are multi-protein intracellular signaling complexes which orchestrate inflammatory responses in immune cells to a diverse range of pathogens and host-derived signals. Widespread inflammasome activation is evident in PD patients at the sites of dopaminergic degeneration as well as in blood samples and mucosal biopsies. Inflammasome activation in the nigrostriatal system is also a common pathological feature in both neurotoxicant and α-synuclein models of PD where dopaminergic degeneration occurs through distinct mechanisms. The NLRP3 (NLR Family Pyrin Domain Containing 3) inflammasome has been shown to be the primary driver of inflammatory neurotoxicity in PD and other neurodegenerative diseases. Chronic NLRP3 inflammasome activation is triggered by pathogenic misfolded α-synuclein aggregates which accumulate and spread over the disease course in PD. Converging lines of evidence suggest that blocking inflammasome activation could be a promising therapeutic strategy for disease modification, with both NLRP3 knockout mice and CNS-permeable pharmacological inhibitors providing robust neuroprotection in multiple PD models. This review summarizes the current evidence and knowledge gaps around inflammasome activation in PD, the pathological mechanisms by which persistent inflammasome activation can drive dopaminergic degeneration and the therapeutic opportunities for disease modification using NLRP3 inhibitors.
Collapse
Affiliation(s)
- Shannon Jewell
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ashane M. Herath
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Richard Gordon
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
50
|
Li W, Jin K, Luo J, Xu W, Wu Y, Zhou J, Wang Y, Xu R, Jiao L, Wang T, Yang G. NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front Cardiovasc Med 2022; 9:988266. [PMID: 36204587 PMCID: PMC9530249 DOI: 10.3389/fcvm.2022.988266] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a common cardiovascular disease with complex pathogenesis, in which multiple pathways and their interweaving regulatory mechanism remain unclear. The primary transcription factor NF-κB plays a critical role in AS via modulating the expression of a series of inflammatory mediators under various stimuli such as cytokines, microbial antigens, and intracellular stresses. Endoplasmic reticulum (ER) stress, caused by the disrupted synthesis and secretion of protein, links inflammation, metabolic signals, and other cellular processes via the unfolded protein response (UPR). Both NF-κB and ER stress share the intersection regarding their molecular regulation and function and are regarded as critical individual contributors to AS. In this review, we summarize the multiple interactions between NF-κB and ER stress activation, including the UPR, NLRP3 inflammasome, and reactive oxygen species (ROS) generation, which have been ignored in the pathogenesis of AS. Given the multiple links between NF-κB and ER stress, we speculate that the integrated network contributes to the understanding of molecular mechanisms of AS. This review aims to provide an insight into these interactions and their underlying roles in the progression of AS, highlighting potential pharmacological targets against the atherosclerotic inflammatory process.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liqun Jiao,
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Tao Wang,
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Tao Wang,
| |
Collapse
|