1
|
Wang J, Liu N, Hu M, Zhang M. Lysosome-related proteins may have changes in the urinary exosomes of patients with acute gout attack. Eur J Med Res 2025; 30:41. [PMID: 39838438 PMCID: PMC11748523 DOI: 10.1186/s40001-025-02272-5] [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: 10/12/2024] [Accepted: 01/01/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The autophagy-lysosome is intricately linked to the development of gout. At present, the diagnosis and monitoring of gout are mainly invasive tests, which cannot predict the occurrence of gout in the acute phase, and bring new pain to patients. This study focuses on the changes of lysosome-related proteins in urinary exosomes of patients with acute gout attack to explore the potential noninvasive biomarkers clinical application value. METHODS Urine samples were collected from the subject and exosomes were extracted. To explore the differentially expressed proteins in urinary exosomes among acute gout patients (AD group), intermittent gout patients (ID group) and normal controls (NC group) by DIA mass spectrometry. Urinary exosomal lysosome associated proteins were analyzed and receiver operating characteristic (ROC) curves of differentially expressed proteins were drawn to evaluate their clinical value in monitoring acute gout attack. RESULTS A total of 1896 proteins were detected between AD group and ID group, of which 121 proteins were differentially expressed (FC > 1.5 and p < 0.05). There were three lysosomal-related proteins differentially expressed in urinary exosomes between AD group and ID group. Compared with the ID group, the expression of Cathepsin Z (CTSZ) and AP-1 complex subunit beta-1 (AP1B1) was increased, while the expression of Lysosome-associated membrane glycoprotein 2 (LAMP2) was decreased in AD group. The ROC analysis showed that CTSZ, AP1B1 and LAMP2 had a strong ability to predict acute gout attack, with AUC of 0.826, 0.847 and 0.882, respectively. CONCLUSIONS There are many specific protein changes in the urinary exosomes of patients with acute gout attack. The urinary exosomes of patients with acute gout attack may exhibit alterations in lysosome-related proteins, particularly CTSZ, AP1B1, and LAMP2, which may become potential biomarkers for monitoring acute gout attack.
Collapse
Affiliation(s)
- Jitu Wang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, 100038, China
| | - Na Liu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, 100038, China
| | - Mei Hu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, 100038, China
| | - Man Zhang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, 100038, China.
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
2
|
Zhang MY, Wang MQ, Huang Y, Gu SL, Zhou MY, Xu ZS, Li LL, Lv M, Cai L, Li R. Silencing aquaporin 1 inhibits autophagy to exert anti-rheumatoid arthritis effects in TNF-α-induced fibroblast-like synoviocytes and adjuvant-induced arthritis rats. Inflamm Res 2025; 74:12. [PMID: 39774992 DOI: 10.1007/s00011-024-01966-6] [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: 10/12/2024] [Revised: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) are key players in rheumatoid arthritis (RA) by resisting apoptosis via increased autophagy. Elevated synovial aquaporin 1 (AQP1) affects RA FLS behaviors, but its relationship with FLS autophagy is unclear. We aim to clarify that silencing AQP1 inhibits autophagy to exert its anti-RA effects. METHODS We studied the effects and mechanisms of AQP1 silencing on autophagy in TNF-α-induced RA FLS and examined the crucial role of autophagy inhibition in its impacts on RA FLS pathogenic behaviors. We explored whether silencing synovial AQP1 relieved rat adjuvant-induced arthritis (AIA) by reducing synovial autophagy. RESULTS TNF-α stimulation increased AQP1 expression and autophagy levels in RA FLS, with a positive correlation between them. AQP1 silencing inhibited autophagy in TNF-α-stimulated RA FLS, along with suppressing proliferation, promoting apoptosis, and mitigating inflammation. Notably, the inhibitory effects of AQP1 silencing on RA FLS pathogenic behaviors were cancelled by autophagy activation with rapamycin (Rapa) but enhanced by autophagy inhibition using 3-Methyladenine. Mechanistically, silencing AQP1 enhanced the binding of Bcl-2 to Beclin1 by decreasing Beclin1-K63 ubiquitination, thus inhibiting RA FLS autophagy. In vivo, silencing synovial AQP1 relieved the severity and development of rat AIA, alongside reducing Ki67 expression, promoting apoptosis, and decreasing autophagy within AIA rat synovium. Expectedly, the Rapa co-administration nullified the anti-AIA effects of silencing synovial AQP1. CONCLUSION These findings reveal that silencing AQP1 inhibits RA FLS pathogenic behaviors and attenuates rat AIA through autophagy inhibition. This study may help clarify the pathogenic role of AQP1 in enhancing autophagy during RA development.
Collapse
Affiliation(s)
- Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Qing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Min Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, China.
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
3
|
Lu X, Zhan T, Zhou Q, Yang W, Liu K, Chen Y, Gao R, Hu J, Gu M, Hu S, Jiao XA, Wang X, Liu X, Liu X. The haemagglutinin-neuraminidase protein of velogenic Newcastle disease virus enhances viral infection through NF-κB-mediated programmed cell death. Vet Res 2024; 55:58. [PMID: 38715081 PMCID: PMC11077864 DOI: 10.1186/s13567-024-01312-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/18/2024] [Indexed: 05/12/2024] Open
Abstract
The haemagglutinin-neuraminidase (HN) protein, a vital membrane glycoprotein, plays a pivotal role in the pathogenesis of Newcastle disease virus (NDV). Previously, we demonstrated that a mutation in the HN protein is essential for the enhanced virulence of JS/7/05/Ch, a velogenic variant NDV strain originating from the mesogenic vaccine strain Mukteswar. Here, we explored the effects of the HN protein during viral infection in vitro using three viruses: JS/7/05/Ch, Mukteswar, and an HN-replacement chimeric NDV, JS/MukHN. Through microscopic observation, CCK-8, and LDH release assays, we demonstrated that compared with Mukteswar and JS/MukHN, JS/7/05/Ch intensified the cellular damage and mortality attributed to the mutant HN protein. Furthermore, JS/7/05/Ch induced greater levels of apoptosis, as evidenced by the activation of caspase-3/8/9. Moreover, JS/7/05/Ch promoted autophagy, leading to increased autophagosome formation and autophagic flux. Subsequent pharmacological experiments revealed that inhibition of apoptosis and autophagy significantly impacted virus replication and cell viability in the JS/7/05/Ch-infected group, whereas less significant effects were observed in the other two infected groups. Notably, the mutant HN protein enhanced JS/7/05/Ch-induced apoptosis and autophagy by suppressing NF-κB activation, while it mitigated the effects of NF-κB on NDV infection. Overall, our study offers novel insights into the mechanisms underlying the increased virulence of NDV and serves as a reference for the development of vaccines.
Collapse
Affiliation(s)
- Xiaolong Lu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Tiansong Zhan
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Qiwen Zhou
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Wenhao Yang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Kaituo Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Ruyi Gao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
| | - Jiao Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China
| | - Xin-An Jiao
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225000, China
| | - Xiaoquan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China.
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225000, China.
| | - Xiaowen Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225000, China.
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225000, China.
| |
Collapse
|
4
|
Nurmi K, Silventoinen K, Keskitalo S, Rajamäki K, Kouri VP, Kinnunen M, Jalil S, Maldonado R, Wartiovaara K, Nievas EI, Denita-Juárez SP, Duncan CJA, Kuismin O, Saarela J, Romo I, Martelius T, Parantainen J, Beklen A, Bilicka M, Matikainen S, Nordström DC, Kaustio M, Wartiovaara-Kautto U, Kilpivaara O, Klein C, Hauck F, Jahkola T, Hautala T, Varjosalo M, Barreto G, Seppänen MRJ, Eklund KK. Truncating NFKB1 variants cause combined NLRP3 inflammasome activation and type I interferon signaling and predispose to necrotizing fasciitis. Cell Rep Med 2024; 5:101503. [PMID: 38593810 PMCID: PMC11031424 DOI: 10.1016/j.xcrm.2024.101503] [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: 12/14/2022] [Revised: 01/04/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
In monogenic autoinflammatory diseases, mutations in genes regulating innate immune responses often lead to uncontrolled activation of inflammasome pathways or the type I interferon (IFN-I) response. We describe a mechanism of autoinflammation potentially predisposing patients to life-threatening necrotizing soft tissue inflammation. Six unrelated families are identified in which affected members present with necrotizing fasciitis or severe soft tissue inflammations. Exome sequencing reveals truncating monoallelic loss-of-function variants of nuclear factor κ light-chain enhancer of activated B cells (NFKB1) in affected patients. In patients' macrophages and in NFKB1-variant-bearing THP-1 cells, activation increases both interleukin (IL)-1β secretion and IFN-I signaling. Truncation of NF-κB1 impairs autophagy, accompanied by the accumulation of reactive oxygen species and reduced degradation of inflammasome receptor nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein 3 (NLRP3), and Toll/IL-1 receptor domain-containing adaptor protein inducing IFN-β (TRIF), thus leading to combined excessive inflammasome and IFN-I activity. Many of the patients respond to anti-inflammatory treatment, and targeting IL-1β and/or IFN-I signaling could represent a therapeutic approach for these patients.
Collapse
Affiliation(s)
- Katariina Nurmi
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Kristiina Silventoinen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Salla Keskitalo
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Kristiina Rajamäki
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Medical and Clinical Genetics, Applied Tumor Genomics Research Program, RPU, UH, 00014 Helsinki, Finland
| | - Vesa-Petteri Kouri
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Matias Kinnunen
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Sami Jalil
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | - Rocio Maldonado
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | - Kirmo Wartiovaara
- Clinical Genetics UH and Helsinki University Hospital (HUH), 00014 Helsinki, Finland
| | | | | | - Christopher J A Duncan
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 4HH, UK
| | - Outi Kuismin
- Department of Clinical Genetics, Oulu University Hospital (OUH), 90014 Oulu, Finland; PEDEGO Research Unit and Medical Research Center Oulu, OUH and University of Oulu (OU), 90014 Oulu, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, HiLIFE, UH, 00014 Helsinki, Finland; Centre for Molecular Medicine Norway, University of Oslo, 0313 Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Inka Romo
- Inflammation Center, Department of Infectious Disease, HUH, 00029 Helsinki, Finland
| | - Timi Martelius
- Inflammation Center, Department of Infectious Disease, HUH, 00029 Helsinki, Finland
| | - Jukka Parantainen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Arzu Beklen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Marcelina Bilicka
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Sampsa Matikainen
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Dan C Nordström
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Internal Medicine and Rehabilitation, HUH and UH, 00029 Helsinki, Finland
| | - Meri Kaustio
- Institute for Molecular Medicine Finland, HiLIFE, UH, 00014 Helsinki, Finland
| | - Ulla Wartiovaara-Kautto
- Department of Hematology, HUH, Comprehensive Cancer Center, UH, 00029 Helsinki, Finland; Applied Tumor Genomics Research Program, RPU, Faculty of Medicine, UH, 00014 Helsinki, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, RPU, Faculty of Medicine, UH, 00014 Helsinki, Finland; Department of Medical and Clinical Genetics/Medicum, Faculty of Medicine, UH, 00014 Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, UH, 00014 Helsinki, Finland; HUS Diagnostic Center, HUSLAB Laboratory of Genetics, HUH, 00029 Helsinki, Finland
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Fabian Hauck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany
| | - Tiina Jahkola
- Department of Plastic Surgery, HUH, 00029 Helsinki, Finland
| | - Timo Hautala
- Research Unit of Internal Medicine and Biomedicine, OU, and Infectious Diseases Clinic, OUH, 90014 Oulu, Finland
| | - Markku Varjosalo
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, Institute of Biotechnology, HiLIFE, UH, 00014 Helsinki, Finland
| | - Goncalo Barreto
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, HUH and UH, 00029 Helsinki, Finland; Rare Disease Center, Children and Adolescents, HUH and UH, 00029 Helsinki, Finland.
| | - Kari K Eklund
- Faculty of Medicine, Clinicum, Translational Immunology Research Program, Research Program Unit (RPU), University of Helsinki (UH), 00014 Helsinki, Finland; Department of Rheumatology, HUH and UH, 00029 Helsinki, Finland; Orton Orthopaedic Hospital, 00280 Helsinki, Finland.
| |
Collapse
|
5
|
Zhang J, Luo Y, Wu B, Huang X, Zhao M, Wu N, Miao J, Li J, Zhu L, Wu D, Shen M. Identifying functional dysregulation of NOD2 variant Q902K in patients with Yao syndrome. Arthritis Res Ther 2024; 26:58. [PMID: 38395960 PMCID: PMC10885518 DOI: 10.1186/s13075-024-03286-w] [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: 10/07/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The study investigated the pathogenesis of Yao syndrome (YAOS), a rare systemic autoinflammatory disease associated with the nucleotide-binding oligomerization domain containing 2 (NOD2) gene variants. METHODS RNA sequencing analyses were used to detect transcriptomic profile changes. Immunoblot and immunohistochemistry were used to examine the NOD2-mediated inflammatory signaling pathways and ELISA was used to detect cytokines. RESULTS Transcriptome analysis of YAOS revealed NOD-like receptor signaling pathway enrichment. Compared with HCs, P-RIP2, p-p65, p-p38, p-ERK, and p-JNK notably increased in PBMCs of a patient with YAOS. P-RIP2, p-p65, and p-p38 elevated in small intestinal mucosa tissues. P-p65 and p-p38 in synovial tissues from YAOS were higher than those in patients with rheumatoid arthritis (RA) and osteoarthritis (OA). Serum interleukin (IL)-6 level along with tumor necrosis factor (TNF)-α and IL-6 secreted from PBMCs were markedly higher in patients with YAOS in comparison to healthy controls (HCs). The supernatants of synovial cells from a patient with YAOS showed substantially higher IL-1β and IL-6 levels than those of RA and OA. Canakinumab therapy of a Q902K heterozygous patient with YAOS resulted in notable clinical improvement. CONCLUSION Overproduction of pro-inflammatory cytokines and the hyperactivation of NOD2-mediated signaling pathways were found in the NOD2 variant Q902K patient with YAOS. NOD2-RIP2-MAPK pathway might play a pivotal role in the pathogenesis of YAOS. These results provide new perspectives for targeted therapies in YAOS.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Yi Luo
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Bingxuan Wu
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Xin Huang
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Mengzhu Zhao
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Na Wu
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Junke Miao
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Ji Li
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Lei Zhu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Di Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Min Shen
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
6
|
Ajayi T, Rai P, Shi M, Gabor KA, Karmaus PWF, Meacham JM, Katen K, Madenspacher JH, Schurman SH, Fessler MB. Race-specific association of an IRGM risk allele with cytokine expression in human subjects. Sci Rep 2023; 13:12911. [PMID: 37558924 PMCID: PMC10412543 DOI: 10.1038/s41598-023-40313-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/08/2023] [Indexed: 08/11/2023] Open
Abstract
Immunity-related GTPase family M (IRGM), located on human chromosome 5q33.1, encodes a protein that promotes autophagy and suppresses the innate immune response. The minor allele of rs13361189 (-4299T>C), a single nucleotide polymorphism in the IRGM promoter, has been associated with several diseases, including Crohn's disease and tuberculosis. Although patterns of linkage disequilibrium and minor allele frequency for this polymorphism differ dramatically between subjects of European and African descent, studies of rs13361189 have predominantly been conducted in Europeans and the mechanism of association is poorly understood. We recruited a cohort of 68 individuals (30 White, 34 African American, 4 other race) with varying rs13361189 genotypes and assessed a panel of immune response measures including whole blood cytokine induction following ex vivo stimulation with Toll-like Receptor ligands. Minor allele carriers were found to have increased serum immunoglobulin M, C-reactive protein, and circulating CD8+ T cells. No differences in whole blood cytokines were observed between minor allele carriers and non-carriers in the overall study population; however, minor allele status was associated with increased induction of a subset of cytokines among African American subjects, and decreased induction among White subjects. These findings underline the importance of broad racial inclusion in genetic studies of immunity.
Collapse
Affiliation(s)
- Teminioluwa Ajayi
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Prashant Rai
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Min Shi
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Kristin A Gabor
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Peer W F Karmaus
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Julie M Meacham
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Kevin Katen
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Jennifer H Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Shepherd H Schurman
- Clinical Research Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
- Clinical Research Unit, National Institute on Aging, Baltimore, MD, 21225, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, MD D2-01, P.O. Box 12233, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
7
|
Hua Y, Wu N, Miao J, Shen M. Single-cell transcriptomic analysis in two patients with rare systemic autoinflammatory diseases treated with anti-TNF therapy. Front Immunol 2023; 14:1091336. [PMID: 36911721 PMCID: PMC9998688 DOI: 10.3389/fimmu.2023.1091336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/08/2023] [Indexed: 02/26/2023] Open
Abstract
Systemic autoinflammatory diseases (SAIDs) are a group of rare diseases characterized by recurrent or continuous inflammation, typically accompanied by genetic variants. Good responses to anti-TNF therapy were observed in SAIDs patients. However, the mechanisms underlying the disease flare and the response to TNF blocking therapy have not been fully elucidated. Here, single-cell RNA sequencing technology was used to describe the transcriptomic profile of PBMCs and PMNs in two SAID patients both before and after anti-TNF treatment. Interferon responses were involved in the disease flare. After anti-TNF therapy, clinical symptoms were alleviated while TNF and IL-1 were unexpectedly increased, indicating that these inflammatory cytokines are not positively correlated with disease activity. Trajectory analysis showed that inhibition of macrophage differentiation, rather than reduction of the inflammatory cytokines, as the potential mechanism of anti-TNF treatment response in SAIDs.
Collapse
Affiliation(s)
- Yichao Hua
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Na Wu
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Junke Miao
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Min Shen
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
8
|
Zhou J, Huang Q, Wang L, Li E, Huang W, Xiang Z. Autophagy Protects Ocular Surface Against Overactivated Inflammation by Degrading Retinoic Acid-Induced Gene-I in Human Conjunctival Epithelial Cells. J Ocul Pharmacol Ther 2022; 38:331-338. [PMID: 35613408 DOI: 10.1089/jop.2021.0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Purpose: To evaluate the pathological role of autophagy in dry eye diseases by detecting the autophagic degradation of RIG-I, a master RNA-sensing receptor in cells. Methods: RNA-sequencing analysis and qPCR analysis of the expression level of genes related to IFN-I signaling pathway was used to evaluate the inflammatory level of cells overexpressed with RIG-I or empty vector, which was further confirmed by WB analysis. Chemical treatment (3-methyladenine, chloroquine, NH4Cl, rapamycin, torin 1 or trehalose) or gene knockdown was used to modulate autophagy. When the autophagy level was regulated, the autophagic degradation of RIG-I and its pathological role in dry eye diseases were determined by detecting the protein level of RIG-I and the level of cell inflammation. Results: Cells that overexpressed RIG-I showed increased expression of genes involved in the IFN-I signaling pathway compared with cells transfected with an empty vector. Inhibition of autophagy leaded to the accumulation of RIG-I in HCECs, combined with the aggravation of the RIG-I-mediated IFN-I signaling pathway. Contrarily, promoting the autophagic degradation of RIG-I by trehalose treatment could alleviate IFN-I signaling pathway. Conclusions: Autophagy could protect the ocular surface against IFN-I signaling pathway by degrading RIG-I in HCECs. This process may restrict the overactivation of inflammation in the pathological development of dry eye disease.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Qinzhu Huang
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Ledan Wang
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Enhui Li
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Wenjuan Huang
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Zhenyang Xiang
- Department of Ophthalmology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| |
Collapse
|
9
|
Yao Q, Shen M, Gorevic P. NOD2 Versus MEFV: Differential Diagnosis of Yao Syndrome and Familial Mediterranean Fever. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:233-239. [PMID: 36467985 PMCID: PMC9524798 DOI: 10.2478/rir-2021-0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/16/2021] [Indexed: 06/17/2023]
Abstract
OBJECTIVES Yao syndrome (YAOS, OMIM 617321) was formerly designated as nucleotide-binding oligomerization domain-containing protein-2 (NOD2)-associated autoinflammatory disease (NAID). This disorder shares similar clinical phenotypes with hereditary periodic fever syndromes (HPFS). This study aimed to compare YAOS with familial Mediterranean fever (FMF). METHODS In this retrospective study, electronic medical records of a case series of YAOS were reviewed and data were analyzed. All patients underwent genetic testing for periodic fever syndrome 6-gene panel. RESULTS A total of 6 cases were presented. These patients were initially thought to have MEditerranean FeVer (MEFV)-negative FMF and received treatment with colchicine. They were eventually diagnosed with YAOS. The differences between these diseases were illustrated. In addition, both MEFV and NOD2 mutations were detected in some patients and family members. Patients with carriage of both gene mutations may present with heterogeneous disease expression. A close correlation between phenotypes and genotypes is needed to make a diagnosis. CONCLUSIONS YAOS may mimic FMF. Molecular analysis should cover NOD2 whole gene sequencing to help distinguish these diseases. Both NOD2 and MEFV mutations may contribute to disease expression in an individual.
Collapse
Affiliation(s)
- Qingping Yao
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, USA
| | - Min Shen
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peter Gorevic
- Department of Rheumatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
10
|
Barrera MJ, Aguilera S, Castro I, Matus S, Carvajal P, Molina C, González S, Jara D, Hermoso M, González MJ. Tofacitinib counteracts IL-6 overexpression induced by deficient autophagy: implications in Sjögren's syndrome. Rheumatology (Oxford) 2021; 60:1951-1962. [PMID: 33216905 DOI: 10.1093/rheumatology/keaa670] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Altered homeostasis of salivary gland (SG) epithelial cells in Sjögren's syndrome (SS) could be the initiating factor that leads to inflammation, secretory dysfunction and autoimmunity. Autophagy is an important homeostatic mechanism, whose deficiency is associated with inflammation and accumulation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) components. We aimed to evaluate whether autophagy is altered in labial SG (LSG) epithelial cells from primary SS (pSS) patients and whether this contributes to inflammation through the JAK-STAT pathway. Furthermore, we investigated the anti-inflammatory effect of the JAK inhibitor tofacitinib in autophagy-deficient (ATG5 knockdown) three-dimensional (3D)-acini. METHODS We analysed LSG biopsies from 12 pSS patients with low focus score and 10 controls. ATG5-deficient 3D-acini were generated and incubated with IL-6 in the presence or absence of tofacitinib. Autophagy markers, pro-inflammatory cytokine expression, and JAK-STAT pathway activation were evaluated by PCR or western blot, along with correlation analyses between the evaluated markers and clinical parameters. RESULTS LSG from pSS patients showed increased p62 and decreased ATG5 expression, correlating negatively with increased activation of JAK-STAT pathway components (pSTAT1 and pSTAT3). Increased expression of STAT1 and IL-6 correlated with EULAR Sjögren's syndrome disease activity index and the presence of anti-Ro antibodies. ATG5-deficient 3D-acini reproduced the findings observed in LSG from pSS patients, showing increased expression of pro-inflammatory markers such as IL-6, which was reversed by tofacitinib. CONCLUSION Decreased expression of ATG5 in LSG epithelial cells from pSS patients possibly contributes to increased inflammation associated with JAK-STAT pathway activation, as evidenced in ATG5-deficient 3D-acini. Interestingly, these results suggest that tofacitinib could be used as an anti-inflammatory agent in pSS patients.
Collapse
Affiliation(s)
| | | | - Isabel Castro
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Soledad Matus
- Fundación Ciencia & Vida, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Patricia Carvajal
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudio Molina
- Facultad de Odontología, Universidad San Sebastián, Santiago, Chile
| | - Sergio González
- Escuela de Odontología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Daniela Jara
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marcela Hermoso
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María-Julieta González
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
11
|
He SD, Huang SG, Zhu HJ, Luo XG, Liao KH, Zhang JY, Tan N, Li DY. Oridonin suppresses autophagy and survival in rheumatoid arthritis fibroblast-like synoviocytes. PHARMACEUTICAL BIOLOGY 2020; 58:146-151. [PMID: 31971852 PMCID: PMC7034059 DOI: 10.1080/13880209.2020.1711783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 06/10/2023]
Abstract
Context: Oridonin exhibits various pharmacological and physiological activities, including antioxidant, antibacterial, anti-inflammatory, pro-apoptotic, anticancer and neurological effects. However, its role in rheumatoid arthritis (RA) is yet to be revealed.Objective: We evaluated the effects of oridonin on the survival and autophagy of RA-fibroblast-like synoviocytes (FLSs).Materials and methods: RA-FLSs were treated with oridonin at serial concentrations of 0, 2, 4, 6, 8 and 10 µg/mL for 24, 48 and 72 h. Then, cell proliferation and apoptosis were measured. A GFP-LC3 plasmid was transfected into the cells to determine autophagy.Results: Oridonin suppressed RA-FLS proliferation in a dose-dependent manner. The half maximal inhibitory concentrations (IC50) of oridonin at 24, 48 and 72 h were 8.28, 7.88 and 8.35 µg/mL, respectively. Treatment with oridonin for 24 h increased apoptosis by 4.1%, and increased the protein levels of Bax and cleaved caspase-3 but significantly decreased the levels of IL-1β in the culture supernatant (p < 0.05). In addition, 6 h of oridonin treatment significantly decreased the number of GFP-LC3 punctate dots and inhibited the protein levels of ATG5 and Beclin1 by 80.01% and 42.12%, respectively. Chloroquine (CQ) significantly reinforced the effects of oridonin on inhibition of autophagy, suppression of proliferation, and induction of apoptosis in RA-FLSs (p < 0.05).Conclusions: Our results indicate that treatment with oridonin in combination with CQ inhibits autophagy and cell proliferation and induces apoptosis in RA-FLSs more effectively than treatment oridonin alone. This finding indicates that oridonin is a potential therapeutic agent for RA.
Collapse
Affiliation(s)
- Shou-Di He
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Sheng-Guang Huang
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Hui-Jun Zhu
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Xiao-Guang Luo
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Kang-Han Liao
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Jie-Yao Zhang
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Ning Tan
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - De-Yu Li
- Traditional Chinese Medicine Department of Rheumatism, Huazhong University of Science and Techology Union Shenzhen Hospital, Shenzhen Nanshan People's Hospital, Shenzhen, China
| |
Collapse
|
12
|
Wang Z, Li C. Xenophagy in innate immunity: A battle between host and pathogen. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 109:103693. [PMID: 32243873 DOI: 10.1016/j.dci.2020.103693] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Autophagy is a fundamental bulk intracellular degradation and recycling process that directly eliminates intracellular microorganisms through "xenophagy" in various types of cells, especially in macrophages. Meanwhile, bacteria have evolved strategies and cellular self-defense mechanisms to prevent autophagosomal degradation and even attack the immune system of host. The lack of knowledge about the roles of autophagy in innate immunity severely limits our understanding of host defensive system and the development of farmed industry consisting of aquaculture. Increasing evidence in recent decades has shown the importance of autophagy. This review focuses on the triggering of xenophagy, targeting of invading pathogens to autophagosomes and elimination in the autophagolysosomes during pathogen infection. How the pathogen can escape from the xenophagy pathway was also discussed. Overall, we aim to reduce diseases and improve industrial production in aquaculture by providing theoretical and technical guidance on xenophagy.
Collapse
Affiliation(s)
- Zhenhui Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
13
|
Haijes HA, Jaeken J, van Hasselt PM. Hypothesis: determining phenotypic specificity facilitates understanding of pathophysiology in rare genetic disorders. J Inherit Metab Dis 2020; 43:701-711. [PMID: 31804708 PMCID: PMC7383723 DOI: 10.1002/jimd.12201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
Abstract
In the rapidly growing group of rare genetic disorders, data scarcity demands an intelligible use of available data, in order to improve understanding of underlying pathophysiology. We hypothesize, based on the principle that clinical similarities may be indicative of shared pathophysiology, that determining phenotypic specificity could provide unsuspected insights in pathophysiology of rare genetic disorders. We explored our hypothesis by studying subunit deficiencies of the conserved oligomeric Golgi (COG) complex, a subgroup of congenital disorders of glycosylation (CDG). In this systematic data assessment, all 45 reported patients with COG-CDG were included. The vocabulary of the Human Phenotype Ontology was used to annotate all phenotypic features and to assess occurrence in other genetic disorders. Gene occurrence ratios were calculated by dividing the frequency in the patient cohort over the number of associated genes, according to the Human Phenotype Ontology. Prioritisation based on phenotypic specificity was highly informative and captured phenotypic features commonly associated with glycosylation disorders. Moreover, it captured features not seen in any other glycosylation disorder, among which episodic fever, likely reflecting underappreciated other cellular functions of the COG complex. Interestingly, the COG complex was recently implicated in the autophagy pathway, as are more than half of the genes underlying disorders that present with episodic fever. This suggests that whereas many phenotypic features in these patients are caused by disrupted glycosylation, episodic fever might be caused by disrupted autophagy. Thus, we here demonstrate support for our hypothesis that determining phenotypic specificity could facilitate understanding of pathophysiology in rare genetic disorders.
Collapse
Affiliation(s)
- Hanneke A. Haijes
- Department of Biomedical Genetics, Section Metabolic DiagnosticsWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Department of Pediatrics, Subdivision Metabolic DiseasesWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jaak Jaeken
- Department of PediatricsCentre for Metabolic Diseases, University Hospital GasthuisbergLeuvenBelgium
| | - Peter M. van Hasselt
- Department of Pediatrics, Subdivision Metabolic DiseasesWilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
14
|
Ruan C, Wang C, Gong X, Zhang Y, Deng W, Zhou J, Huang D, Wang Z, Zhang Q, Guo A, Lu J, Gao J, Peng D, Xue Y. An integrative multi-omics approach uncovers the regulatory role of CDK7 and CDK4 in autophagy activation induced by silica nanoparticles. Autophagy 2020; 17:1426-1447. [PMID: 32397800 DOI: 10.1080/15548627.2020.1763019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysfunction of macroautophagy/autophagy has been postulated as a major cellular toxicological response to nanomaterials. It has been reported that excessive autophagy activation, induced by silica nanoparticles (SiNPs), contributes to autophagy dysfunction, whereas little is known how SiNPs trigger autophagy activation. Here, we treated normal rat kidney (NRK) cells using 3 different sizes of SiNPs (16, 29, and 51 nm) and observed that 16-nm SiNPs, with a final concentration of 60 μg/mL, dramatically induce autophagy activation without reducing cell viability. We further conducted a transcriptomic, proteomic, and phosphoproteomic profiling, and detected 23 autophagy-related (Atg) genes and 35 autophagy regulators regulated on at least one omic layer. To identify key regulators from the multi-omics data, we developed a new algorithm of computational prediction of master autophagy-regulating kinases (cMAK) to detect 21 candidates and revealed the CDK7-CDK4 cascade to be functional. The silence or inhibition of Cdk7 or Cdk4 significantly attenuated autophagic activation but not influenced autophagic flux blockage induced by 16-nm SiNPs. Further computational modeling indicated that the CDK7-CDK4 signaling axis potentially triggers autophagy activation by phosphorylating RB1 (RB transcriptional corepressor 1), activating two critical transcription factors, E2F1 (E2F transcription factor 1) and FOXO3 (forkhead box O3), and enhancing the transcriptional levels of at least 8 Atg genes and autophagy regulators in response to SiNPs. Our studies not only established a powerful method for predicting regulatory kinases from the multi-omics data but also revealed a potential mechanism of SiNP-triggered autophagy activation through modulating the CDK7-CDK4 cascade.Abbreviations: 3-MA: 3-methyladenine; Atg: autophagy-related; BECN1: beclin 1; CCK-8: cell counting kit-8; CDK4: cyclin dependent kinase 4; CDK7: cyclin dependent kinase 7; cMAK: computational prediction of master autophagy-regulating kinases; CQ: chloroquine; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; E-ratio: enrichment ratio; E2F1: E2F transcription factor 1; EBSS: Earle's balanced salt solution; ER: endoplasmic reticulum; FOXO3: forkhead box O3; FPKM: fragments per kilobase of exon per million fragments mapped; GO: gene ontology; H2O2: hydrogen peroxide; iGPS: in vivo GPS; KEGG: Kyoto Encyclopedia of Genes and Genomes; LC-MS/MS: liquid chromatography-tandem mass spectrometry; LDH: lactate dehydrogenase; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; NRK: normal rat kidney; p-site: phosphorylation site; PBS: phosphate-buffered saline; PDI: polydispersity index; PTM: post-translational modification; QKS: quantitative kinase state; RB1: RB transcriptional corepressor 1; RBHs: reciprocal best hits; RNA-Seq: RNA sequencing; ROS: reactive oxygen species; rSiNPs: SiNPs fluorescently labeled with rhodamine B; SEM: scanning electronic microscopy; SiNPs: silica nanoparticles; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; ssKSR: site-specific kinase-substrate relation; TEM: transmission electron microscopy; tfLC3: mRFP-GFP tandem fluorescent-tagged LC3.
Collapse
Affiliation(s)
- Chen Ruan
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Chenwei Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xuanqing Gong
- State Key Laboratory of Physical Chemistry of Solid Surfaces, the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Ying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wankun Deng
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Zhou
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Dengtong Huang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Zining Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qiong Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Anyuan Guo
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, SAR
| | - Jinhao Gao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Di Peng
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xue
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Hua Y, Wu D, Shen M, Yu K, Zhang W, Zeng X. Phenotypes and genotypes of Chinese adult patients with systemic autoinflammatory diseases. Semin Arthritis Rheum 2019; 49:446-452. [DOI: 10.1016/j.semarthrit.2019.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/27/2019] [Accepted: 05/06/2019] [Indexed: 11/15/2022]
|
16
|
Tsuji S, Matsuzaki H, Iseki M, Nagasu A, Hirano H, Ishihara K, Ueda N, Honda Y, Horiuchi T, Nishikomori R, Morita Y, Mukai T. Functional analysis of a novel G87V TNFRSF1A mutation in patients with TNF receptor-associated periodic syndrome. Clin Exp Immunol 2019; 198:416-429. [PMID: 31429073 DOI: 10.1111/cei.13365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2019] [Indexed: 12/17/2022] Open
Abstract
Tumor necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is an autoinflammatory disease that is caused by heterozygous mutations in the TNFRSF1A gene. Although more than 150 TNFRSF1A mutations have been reported to be associated with TRAPS phenotypes only a few, such as p.Thr79Met (T79M) and cysteine mutations, have been functionally analyzed. We identified two TRAPS patients in one family harboring a novel p.Gly87Val (G87V) mutation in addition to a p.Thr90Ile (T90I) mutation in TNFRSF1A. In this study, we examined the functional features of this novel G87V mutation. In-vitro analyses using mutant TNF receptor 1 (TNF-R1)-over-expressing cells demonstrated that this mutation alters the expression and function of TNF-R1 similar to that with the previously identified pathogenic T79M mutation. Specifically, cell surface expression of the mutant TNF-R1 in transfected cells was inhibited with both G87V and T79M mutations, whereas the T90I mutation did not affect this. Moreover, peripheral blood mononuclear cells (PBMCs) from TRAPS patients harboring the G87V and T90I mutations showed increased mitochondrial reactive oxygen species (ROS). Furthermore, the effect of various Toll-like receptor (TLR) ligands on inflammatory responses was explored, revealing that PBMCs from TRAPS patients are hyper-responsive to TLR-2 and TLR-4 ligands and that interleukin (IL)-8 and granulocyte-macrophage colony-stimulating factor (GM-CSF) are likely to be involved in the pathogenesis of TRAPS. These findings suggest that the newly identified G87V mutation is one of the causative mutations of TRAPS. Our findings based on unique TRAPS-associated mutations provide novel insight for clearer understanding of inflammatory responses, which would be basic findings of developing a new therapeutic and prophylactic approach to TRAPS.
Collapse
Affiliation(s)
- S Tsuji
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - H Matsuzaki
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Hiroshima, Japan
| | - M Iseki
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - A Nagasu
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - H Hirano
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - K Ishihara
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - N Ueda
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Y Honda
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - T Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu, Oita, Japan
| | - R Nishikomori
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Y Morita
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - T Mukai
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| |
Collapse
|
17
|
Methylomic correlates of autophagy activity in cystic fibrosis. J Cyst Fibros 2019; 18:491-500. [PMID: 30737168 DOI: 10.1016/j.jcf.2019.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
Autophagy is a highly regulated, biological process that provides energy during periods of stress and starvation. This conserved process also acts as a defense mechanism and clears microbes from the host cell. Autophagy is impaired in Cystic Fibrosis (CF) patients and CF mice, as their cells exhibit low expression levels of essential autophagy molecules. The genetic disorder in CF is due to mutations in the cystic fibrosis transmembrane conductance regulator (cftr) gene that encodes for a chloride channel. CF patients are particularly prone to infection by pathogens that are otherwise cleared by autophagy in healthy immune cells including Burkholderia cenocepacia (B. cenocepacia). The objective of this study is to determine the mechanism underlying weak autophagic activity in CF macrophages and find therapeutic targets to correct it. Using reduced representation bisulfite sequencing (RRBS) to determine DNA methylation profile, we found that the promoter regions of Atg12 in CF macrophages are significantly more methylated than in the wild-type (WT) immune cells, accompanied by low protein expression. The natural product epigallocatechin-3-gallate (EGCG) significantly reduced the methylation of Atg12 promoter improving its expression. Accordingly, EGCG restricted B. cenocepacia replication within CF mice and their derived macrophages by improving autophagy and preventing dissemination. In addition, EGCG improved the function of CFTR protein. Altogether, utilizing RRBS for the first time in the CF field revealed a previously unrecognized mechanism for reduced autophagic activity in CF. Our data also offers a mechanism by which EGCG exerts its positive effects in CF.
Collapse
|
18
|
Wang L, Ye X, Zhao T. The physiological roles of autophagy in the mammalian life cycle. Biol Rev Camb Philos Soc 2018; 94:503-516. [PMID: 30239126 PMCID: PMC7379196 DOI: 10.1111/brv.12464] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
Abstract
Autophagy is primarily an efficient intracellular catabolic pathway used for degradation of abnormal cellular protein aggregates and damaged organelles. Although autophagy was initially proposed to be a cellular stress responder, increasing evidence suggests that it carries out normal physiological roles in multiple biological processes. To date, autophagy has been identified in most organs and at many different developmental stages, indicating that it is not only essential for cellular homeostasis and renovation, but is also important for organ development. Herein, we summarize our current understanding of the functions of autophagy (which here refers to macroautophagy) in the mammalian life cycle.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Xiongjun Ye
- Department of Urology, Peking University People's Hospital, 100034 Beijing, China
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, 100049 Beijing, China
| |
Collapse
|
19
|
Mechanism of cyclosporine A nephrotoxicity: Oxidative stress, autophagy, and signalings. Food Chem Toxicol 2018; 118:889-907. [DOI: 10.1016/j.fct.2018.06.054] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022]
|