1
|
Sun K, Wu F, Zheng J, Wang H, Li H, Xie Z. Essential blood molecular signature for progression of sepsis-induced acute lung injury: Integrated bioinformatic, single-cell RNA Seq and machine learning analysis. Int J Biol Macromol 2024; 282:136961. [PMID: 39481313 DOI: 10.1016/j.ijbiomac.2024.136961] [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: 08/29/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
In this study, we aimed to identify an essential blood molecular signature for chacterizing the progression of sepsis-induced acute lung injury using integrated bioinformatic and machine learning analysis. The results showed that a total of 88 functionally related ALI-associated hub genes in sepsis were identified by MCODE analysis and they were enriched in infection and inflammtory responses, lung and cardiovascular disease pathways. These hub genes stratified ALI-sepsis and sepsis and further stratified two subtypes of sepsis-ALI with differential ALI scores, hub gene expression patterns, and levels of immune cells. A seven-gene signature including TNFRSF1A, NFKB1, FCGR2A, NFE2L2, ICAM1 and SOCS3 and PDCD1 was derived from the hub genes. These genes were significantly implicated in immune and metabolism pathways. They were expressed in six circulatory immune cells based on analysis of a single cell RNA sequencing dataset. Furthermore, the seven-gene signature was corrobarated using by integrating 12 machine learning algorithms. A premium three-gene signature NFE2L2, FCGR2A and PDCD1 for differentiating ALI-sepsis from sepsis were also derived from the seven-gene signature based on analysis of the seven core hub genes by the machine learning algorithms. Furthermore, the expressions of hub genes were verified in sepsis mice models. Therefore, our study provided an avenue to develop a molecular tool for identify and characterize progression of acute lung injury associated with sepsis.
Collapse
Affiliation(s)
- Keyu Sun
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Fupeng Wu
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Jiayi Zheng
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Han Wang
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Haidong Li
- Research and Translational Laboratory of Acute Injury and Secondary Infection, Minhang Hospital, Fudan University, Shanghai 201199, China.
| | - Zichen Xie
- Emergency Department, Minhang Hospital, Fudan University, Shanghai 201100, China.
| |
Collapse
|
2
|
Xu Y, Yang Z, Wang T, Hu L, Jiao S, Zhou J, Dai T, Feng Z, Li S, Meng Q. From molecular subgroups to molecular targeted therapy in rheumatoid arthritis: A bioinformatics approach. Heliyon 2024; 10:e35774. [PMID: 39220908 PMCID: PMC11365346 DOI: 10.1016/j.heliyon.2024.e35774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
1Background Rheumatoid Arthritis (RA) is a heterogeneous autoimmune disease with multiple unidentified pathogenic factors. The inconsistency between molecular subgroups poses challenges for early diagnosis and personalized treatment strategies. In this study, we aimed to accurately distinguish RA patients at the transcriptome level using bioinformatics methods. 2Methods We collected a total of 362 transcriptome datasets from RA patients in three independent samples from the GEO database. Consensus clustering was performed to identify molecular subgroups, and clinical features were assessed. Differential analysis was employed to annotate the biological functions of specifically upregulated genes between subgroups. 3Results Based on consensus clustering of RA samples, we identified three robust molecular subgroups, with Subgroup III representing the high-risk subgroup and Subgroup II exhibiting a milder phenotype, possibly associated with relatively higher levels of autophagic ability. Subgroup I showed biological functions mainly related to viral infections, cellular metabolism, protein synthesis, and inflammatory responses. Subgroup II involved autophagy of mitochondria and organelles, protein localization, and organelle disassembly pathways, suggesting heterogeneity in the autophagy process of mitochondria that may play a protective role in inflammatory diseases. Subgroup III represented a high-risk subgroup with pathological processes including abnormal amyloid precursor protein activation, promotion of inflammatory response, and cell proliferation. 4Conclusion The classification of the RA dataset revealed pathological heterogeneity among different subgroups, providing new insights and a basis for understanding the molecular mechanisms of RA, identifying potential therapeutic targets, and developing personalized treatment approaches.
Collapse
Affiliation(s)
- Yangyang Xu
- Guizhou Medical University, Guiyang City, Guizhou Province, China
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhenyu Yang
- Jinan University, Guangzhou, Guangdong Province, China
- Xuzhou New Health Hospital, North Hospital of Xuzhou Cancer Hospital, Xuzhou City, Jiangsu Province, China
| | - Tengyan Wang
- Guizhou Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guiyang City, Guizhou Province, China
| | - Liqiong Hu
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| | - Songsong Jiao
- Jinan University, Guangzhou, Guangdong Province, China
| | - Jiangfei Zhou
- Jinan University, Guangzhou, Guangdong Province, China
| | - Tianming Dai
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhencheng Feng
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| | - Siming Li
- Guizhou Medical University, Guiyang City, Guizhou Province, China
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| | - Qinqqi Meng
- Guangzhou Red Cross Hospital Affiliated of Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Efstathiou C, Zhang Y, Kandwal S, Fayne D, Molloy EJ, Stevenson NJ. Respiratory syncytial virus NS1 inhibits anti-viral Interferon-α-induced JAK/STAT signaling, by limiting the nuclear translocation of STAT1. Front Immunol 2024; 15:1395809. [PMID: 38938568 PMCID: PMC11208467 DOI: 10.3389/fimmu.2024.1395809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 06/29/2024] Open
Abstract
Human respiratory viruses are the most prevalent cause of disease in humans, with the highly infectious RSV being the leading cause of infant bronchiolitis and viral pneumonia. Responses to type I IFNs are the primary defense against viral infection. However, RSV proteins have been shown to antagonize type I IFN-mediated antiviral innate immunity, specifically dampening intracellular IFN signaling. Respiratory epithelial cells are the main target for RSV infection. In this study, we found RSV-NS1 interfered with the IFN-α JAK/STAT signaling pathway of epithelial cells. RSV-NS1 expression significantly enhanced IFN-α-mediated phosphorylation of STAT1, but not pSTAT2; and neither STAT1 nor STAT2 total protein levels were affected by RSV-NS1. However, expression of RSV-NS1 significantly reduced ISRE and GAS promoter activity and anti-viral IRG expression. Further mechanistic studies demonstrated RSV-NS1 bound STAT1, with protein modeling indicating a possible interaction site between STAT1 and RSV-NS1. Nuclear translocation of STAT1 was reduced in the presence of RSV-NS1. Additionally, STAT1's interaction with the nuclear transport adapter protein, KPNA1, was also reduced, suggesting a mechanism by which RSV blocks STAT1 nuclear translocation. Indeed, reducing STAT1's access to the nucleus may explain RSV's suppression of IFN JAK/STAT promoter activation and antiviral gene induction. Taken together these results describe a novel mechanism by which RSV controls antiviral IFN-α JAK/STAT responses, which enhances our understanding of RSV's respiratory disease progression.
Collapse
Affiliation(s)
- Claudia Efstathiou
- Viral Immunology Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Yamei Zhang
- Viral Immunology Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Shubhangi Kandwal
- Molecular Design Group, School of Chemical Sciences, Dublin City University, Glasnevin, Ireland
- Molecular Design Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Darren Fayne
- Molecular Design Group, School of Chemical Sciences, Dublin City University, Glasnevin, Ireland
- DCU Life Sciences Institute, Dublin City University, Dublin, Ireland
| | - Eleanor J. Molloy
- Paediatrics, Trinity College, Dublin, Ireland
- Neonatology, Children’s Hospital Ireland at Tallaght, Dublin, Ireland
- Neonatology, Coombe Women’s and Infants University Hospital, Dublin, Ireland
| | - Nigel J. Stevenson
- Viral Immunology Group, Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Eltahir HM, Elbadawy HM, Almikhlafi MA, Alalawi AM, Aldhafiri AJ, Alahmadi YM, Al thagfan SS, Albadrani M, M Eweda S, Abouzied MM. Sitagliptin ameliorates L-arginine-induced acute pancreatitis via modulating inflammatory cytokines expression and combating oxidative stress. Front Pharmacol 2024; 15:1389670. [PMID: 38910880 PMCID: PMC11190672 DOI: 10.3389/fphar.2024.1389670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/26/2024] [Indexed: 06/25/2024] Open
Abstract
Background Acute pancreatitis (AP) is an inflammatory condition that resolves spontaneously, but occasionally, develops into systemic inflammation, organ failure and mortality. Oxidative stress and activation of inflammatory pathways represent major players in AP pathogenesis. Current management of AP relies on attenuating injuries to the pancreas and putting the inflammatory process under control. In this study, we investigated the role of sitagliptin in modulating L-arginine-induced AP in rats. Methods Swiss rats were subdivided into a healthy control group, AP group (a single dose of L-arginine 250 mg/100 g, intraperitoneal), and sitagliptin + L-arginine-treated group (10 mg sitagliptin/kg body weight/day, orally). Sitagliptin treatment started 1 hour after L-arginine injection and continued for 3days. Biochemical and histopathological investigations were performed on serum and tissue samples collected from test animals. Results L-arginine increased pancreatic meyloperoxidase and serum amylase- and lipase activities and serum levels of TNF-α, LT-α, IFN-γ, IL-1α/β, IL-6, IL-10, IL-12, and IL-15. AP animals showed elevated MDA and NO and decreased GSH and serum calcium levels. Histopathological changes were observed by H&E staining. Sitagliptin treatment significantly ameliorated these biochemical and histological changes diminishing the signs of AP. Conclusion Sitagliptin treatment was effective in ameliorating L-arginine-induced AP which can be regarded to its anti-inflammatory and antioxidant effect.
Collapse
Affiliation(s)
- Heba M. Eltahir
- Department of Pharmacology and Toxicology (Biochemistry Subdivision), College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Hossein M. Elbadawy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Mohannad A. Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ali M. Alalawi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ahmed J. Aldhafiri
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Yaser M. Alahmadi
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Sultan S. Al thagfan
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Muayad Albadrani
- Department of Family and Community Medicine, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Saber M Eweda
- 5Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mekky M. Abouzied
- Department of Pharmacology and Toxicology (Biochemistry Subdivision), College of Pharmacy, Taibah University, Medina, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
5
|
Jeong J, Lee J, Talaia G, Kim W, Song J, Hong J, Yoo K, Gonzalez DG, Athonvarangkul D, Shin J, Dann P, Haberman AM, Kim LK, Ferguson SM, Choi J, Wysolmerski J. Intracellular calcium links milk stasis to lysosome-dependent cell death during early mammary gland involution. Cell Mol Life Sci 2024; 81:29. [PMID: 38212474 PMCID: PMC10784359 DOI: 10.1007/s00018-023-05044-8] [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: 06/10/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 01/13/2024]
Abstract
Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 h of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6, and TGFβ3, all of which appear to be upregulated by increased intracellular calcium. We further demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis through a process involving inhibition of CDK4/6 and cell cycle progression. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gabriel Talaia
- Departments of Cell Biology and of Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Wonnam Kim
- Division of Phamacology, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Junho Song
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Juhyeon Hong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kwangmin Yoo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Diana Athonvarangkul
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jaehun Shin
- Integrated Science Engineering Division, Underwood International College, Yonsei University, Seoul, Republic of Korea
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ann M Haberman
- Departments of Immunobiology and Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06230, Republic of Korea
| | - Shawn M Ferguson
- Departments of Cell Biology and of Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - John Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
6
|
Luo P, Ji Y, Liu X, Zhang W, Cheng R, Zhang S, Qian X, Huang C. Affected inflammation-related signaling pathways in snake envenomation: A recent insight. Toxicon 2023; 234:107288. [PMID: 37703930 DOI: 10.1016/j.toxicon.2023.107288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
Snake envenomation is well known to cause grievous pathological signs, including haemorrhagic discharge, necrosis, and respiratory distress. However, inflammatory reactions are also common envenoming manifestations that lead to successive damage, such as oedema, ulceration, lymphadenectasis, systemic inflammatory response syndrome (SIRS) and even multiple organ dysfunction syndrome (MODS). Interference with the inflammatory burst is hence important in the clinical treatment of snake envenomation. Here, we summarize the typical snake toxins (or venoms) that cause inflammatory reactions and the underlying signaling pathways. In brief, inflammatory reactions are usually triggered by snake venom phospholipase A2 (svPLA2), snake venom metalloprotease (SVMP), snake venom serine protease (SVSP) and C-type lectin/snaclec (CTL) as well as disintegrin (DIS) via multiple signaling pathways. They are nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3), nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), janus kinase/signal transducer and activator of transcription (JAK-STAT) and phosphoinositide 3-Kinase/protein kinase B (PI3K/PKB also called PI3K-AKT) signaling pathways. Activation of these pathways promotes the expression of pro-inflammatory molecules such as cytokines, especially interleukin-1β (IL-1β) which causes further inflammatory cascades and manifestations, such as swelling, fever, pain, and severe complications. Remarkably, almost half of introduced snake toxins (or venoms) have anti-inflammatory effects through blocking these pathways and suppressing the expression of pro-inflammatory molecules. Investigation of affected inflammation-related signaling pathways is meaningful to achieve better clinical treatment.
Collapse
Affiliation(s)
- Peiyi Luo
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Yuxin Ji
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiaohan Liu
- Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China.
| | - Weiyun Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Ruoxi Cheng
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Shuxian Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiao Qian
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Chunhong Huang
- College of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| |
Collapse
|
7
|
Wang Q, Shi Q, Wang Z, Lu J, Hou J. Integrating plasma proteomes with genome-wide association data for causal protein identification in multiple myeloma. BMC Med 2023; 21:377. [PMID: 37775746 PMCID: PMC10542236 DOI: 10.1186/s12916-023-03086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a severely debilitating and fatal B-cell neoplastic disease. The discovery of disease-associated proteins with causal genetic evidence offers a chance to uncover novel therapeutic targets. METHODS First, we comprehensively investigated the causal association between 2994 proteins and MM through two-sample mendelian randomization (MR) analysis using summary-level data from public genome-wide association studies of plasma proteome (N = 3301 healthy individuals) and MM (598 cases and 180,756 controls). Sensitivity analyses were performed for these identified causal proteins. Furthermore, we pursued the exploration of enriched biological pathways, prioritized the therapeutic proteins, and evaluated their druggability using the KEGG pathway analysis, MR-Bayesian model averaging analysis, and cross-reference with current databases, respectively. RESULTS We identified 13 proteins causally associated with MM risk (false discovery rate corrected P < 0.05). Six proteins were positively associated with the risk of MM, including nicotinamide phosphoribosyl transferase (NAMPT; OR [95% CI]: 1.35 [1.18, 1.55]), tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1; 1.14 [1.06, 1.22]), neutrophil cytosol factor 2 (NCF2; 1.27 [1.12, 1.44]), carbonyl reductase 1, cAMP-specific 3',5'-cyclic phosphodiesterase 4D (PDE4D), platelet-activating factor acetylhydrolase IB subunit beta (PAFAH1B2). Seven proteins were inversely associated with MM, which referred to suppressor of cytokine signaling 3 (SOCS3; 0.90 [0.86, 0.94]), Fc-gamma receptor III-B (FCGR3B; 0.75 [0.65,0.86]), glypican-1 (GPC1; 0.69 [0.58,0.83]), follistatin-related protein 1, protein tyrosine phosphatase non-receptor type 4 (PTPN4), granzyme B, complement C1q subcomponent subunit C (C1QC). Three of the causal proteins, SOCS3, FCGR3B, and NCF2, were enriched in the osteoclast differentiation pathway in KEGG enrichment analyses while GPC1 (marginal inclusion probability (MIP):0.993; model averaged causal effects (MACE): - 0.349), NAMPT (MIP:0.433; MACE: - 0.113), and NCF2 (MIP:0.324; MACE:0.066) ranked among the top three MM-associated proteins according to MR-BMA analyses. Furthermore, therapeutics targeting four proteins are currently under evaluation, five are druggable and four are future breakthrough points. CONCLUSIONS Our analysis revealed a set of 13 novel proteins, including six risk and seven protective proteins, causally linked to MM risk. The discovery of these MM-associated proteins opens up the possibility for identifying novel therapeutic targets, further advancing the integration of genome and proteome data for drug development.
Collapse
Affiliation(s)
- Qiangsheng Wang
- Department of Hematology, Ningbo Hangzhou Bay Hospital, Ningbo, 315000, Zhejiang, China
| | - Qiqin Shi
- Department of Ophthalmology, Ningbo Hangzhou Bay Hospital, Ningbo, 315000, Zhejiang, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jiawen Lu
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
8
|
Jeong J, Lee J, Talaia G, Kim W, Song J, Hong J, Yoo K, Gonzalez D, Athonvarangkul D, Shin J, Dann P, Haberman A, Kim LK, Ferguson S, Choi J, Wysolmerski J. Intracellular Calcium links Milk Stasis to Lysosome Dependent Cell Death by Activating a TGFβ3/TFEB/STAT3 Pathway Early during Mammary Gland Involution. RESEARCH SQUARE 2023:rs.3.rs-3030763. [PMID: 37398309 PMCID: PMC10312953 DOI: 10.21203/rs.3.rs-3030763/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 hours of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6 and TGFβ3, all of which appear to be upregulated by increased intracellular calcium. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis. This is the result of increased TGFβ signaling and inhibition of cell cycle progression. Finally, we demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3, a process which also appears to be mediated by TGFβ signaling. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Yale School of Medicine: Yale University School of Medicine
| | | | - Gabriel Talaia
- Yale School of Medicine: Yale University School of Medicine
| | | | | | | | | | - David Gonzalez
- Yale School of Medicine: Yale University School of Medicine
| | | | | | - Pamela Dann
- Yale School of Medicine: Yale University School of Medicine
| | - Ann Haberman
- Yale School of Medicine: Yale University School of Medicine
| | | | - Shawn Ferguson
- Yale School of Medicine: Yale University School of Medicine
| | | | | |
Collapse
|
9
|
Dai L, Han Y, Yang Z, Zeng Y, Liang W, Shi Z, Tao Y, Liang X, Liu W, Zhou S, Xing Z, Hu W, Wang X. Identification and validation of SOCS1/2/3/4 as potential prognostic biomarkers and correlate with immune infiltration in glioblastoma. J Cell Mol Med 2023. [PMID: 37315184 PMCID: PMC10399539 DOI: 10.1111/jcmm.17807] [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: 02/06/2023] [Revised: 05/11/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023] Open
Abstract
Suppressor of cytokine signalling (SOCS) 1/2/3/4 are involved in the occurrence and progression of multiple malignancies; however, their prognostic and developmental value in patients with glioblastoma (GBM) remains unclear. The present study used TCGA, ONCOMINE, SangerBox3.0, UALCAN, TIMER2.0, GENEMANIA, TISDB, The Human Protein Atlas (HPA) and other databases to analyse the expression profile, clinical value and prognosis of SOCS1/2/3/4 in GBM, and to explore the potential development mechanism of action of SOCS1/2/3/4 in GBM. The majority of analyses showed that SOCS1/2/3/4 transcription and translation levels in GBM tissues were significantly higher than those in normal tissues. qRT-PCR, western blotting (WB) and immunohistochemical staining were used to verify that SOCS3 was expressed at higher mRNA and protein levels in GBM than in normal tissues or cells. High SOCS1/2/3/4 mRNA expression was associated with poor prognosis in patients with GBM, especially SOCS3. SOCS1/2/3/4 were highly contraindicated, which had few mutations, and were not associated with clinical prognosis. Furthermore, SOCS1/2/3/4 were associated with the infiltration of specific immune cell types. In addition, SOCS3 may affect the prognosis of patients with GBM through JAK/STAT signalling pathway. Analysis of the GBM-specific protein interaction (PPI) network showed that SOCS1/2/3/4 were involved in multiple potential carcinogenic mechanisms of GBM. In addition, colony formation, Transwell, wound healing and western blotting assays revealed that inhibition of SOCS3 decreased the proliferation, migration and invasion of GBM cells. In conclusion, the present study elucidated the expression profile and prognostic value of SOCS1/2/3/4 in GBM, which may provide potential prognostic biomarkers and therapeutic targets for GBM, especially SOCS3.
Collapse
Affiliation(s)
- Lirui Dai
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Yongjie Han
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Zhuo Yang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Yuling Zeng
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wulong Liang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Zimin Shi
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Yiran Tao
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Xianyin Liang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Wanqing Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Weihua Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| |
Collapse
|
10
|
Yang Z, Wei J, He Y, Ren L, Chen S, Deng Y, Zang N, Liu E. Identification of functional pathways and potential genes associated with interferon signaling during human adenovirus type 7 infection by weighted gene coexpression network analysis. Arch Virol 2023; 168:130. [PMID: 37017816 PMCID: PMC10076410 DOI: 10.1007/s00705-023-05707-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/15/2022] [Indexed: 04/06/2023]
Abstract
Human adenovirus type 7 (HAdV-7) can cause severe pneumonia and complications in children. However, the mechanism of pathogenesis and the genes involved remain largely unknown. We collected HAdV-7-infected and mock-infected A549 cells at 24, 48, and 72 hours postinfection (hpi) for RNA sequencing (RNA-Seq) and identified potential genes and functional pathways associated with HAdV-7 infection using weighted gene coexpression network analysis (WGCNA). Based on bioinformatics analysis, 12 coexpression modules were constructed by WGCNA, with the blue, tan, and brown modules significantly positively correlated with adenovirus infection at 24, 48, and 72 hpi, respectively. Functional enrichment analysis indicated that the blue module was mainly enriched in DNA replication and viral processes, the tan module was largely enriched in metabolic pathways and regulation of superoxide radical removal, and the brown module was predominantly enriched in regulation of cell death. qPCR was used to determine transcript abundance of some identified hub genes, and the results were consistent with those from RNA-Seq. Comprehensively analyzing hub genes and differentially expressed genes in the GSE68004 dataset, we identified SOCS3, OASL, ISG15, and IFIT1 as potential candidate genes for use as biomarkers or drug targets in HAdV-7 infection. We propose a multi-target inhibition of the interferon signaling mechanism to explain the association of HAdV-7 infection with the severity of clinical consequences. This study has allowed us to construct a framework of coexpression gene modules in A549 cells infected with HAdV-7, thus providing a basis for identifying potential genes and pathways involved in adenovirus infection and for investigating the pathogenesis of adenovirus-associated diseases.
Collapse
Affiliation(s)
- Zhongying Yang
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jianhua Wei
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yu He
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Luo Ren
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Shiyi Chen
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yu Deng
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Na Zang
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Enmei Liu
- Department of Respiratory Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders,Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| |
Collapse
|
11
|
Emamgholipour S, Esmaeili F, Shabani M, Hasanpour SZ, Pilehvari M, Zabihi-Mahmoudabadi H, Motevasseli M, Shanaki M. Alterations of SOCS1 and SOCS3 transcript levels, but not promoter methylation levels in subcutaneous adipose tissues in obese women. BMC Endocr Disord 2023; 23:7. [PMID: 36609306 PMCID: PMC9817302 DOI: 10.1186/s12902-022-01247-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Animal model studies suggest that change in the members of the suppressor of the cytokine signaling (SOCS) family (mainly SOCS1 and SOCS3) is linked to the pathogenesis of obesity-related metabolic disorders. Moreover, epigenetic modification is involved in the transcriptional regulation of the SOCS gene family. Here, we aimed to evaluate the mRNA expression as well as gene promoter methylation of SOCS1 and SOCS3 in subcutaneous adipose tissue (SAT) from obese women compared to normal-weight subjects. We also intend to identify the possible association of SOCS1 and SOCS3 transcript levels with metabolic parameters in the context of obesity. METHODS This study was conducted on women with obesity (n = 24) [body mass index (BMI) ≥ 30 kg/m 2] and women with normal-weight (n = 22) (BMI < 25 kg/m 2). Transcript levels of SOCS1 and SOCS3 were evaluated by real-time PCR in SAT from all participants. After bisulfite treatment of DNA, methylation-specific PCR was used to assess the putative methylation of 10 CpG sites in the promoter of SOCS1 and 13 CpG sites in SOCS3 in SAT from women with obesity and normal weight. RESULTS It was found that unlike SOCS3, which disclosed an elevating expression pattern, the expression level of SOCS1 was lower in the women with obesity as compared with their non-obese counterparts (P-value = 0.03 for SOCS1 transcript level and P-value = 0.011 for SOCS3 transcript level). As for the analysis of promoter methylation, it was found that SOCS1 and SOCS3 methylation were not significantly different between the individuals with obesity and normal weight (P-value = 0.45 and P-value = 0.89). Correlation analysis indicated that the transcript level of SOCS1 mRNA expression had an inverse correlation with BMI, hs-CRP levels, HOMA-IR, and insulin levels. However, the SOCS3 transcript level showed a positive correlation with BMI, waist-to-height ratio, waist circumference, hip circumference, hs-CRP, HOMA-IR, insulin, fasting blood glucose, and total cholesterol. Interestingly, HOMA-IR is the predictor of the transcript level of SOCS1 (β = - 0.448, P-value = 0.003) and SOCS3 (β = 0.465, P-value = 0.002) in SAT of all participants. CONCLUSIONS Our findings point to alterations of SOCS1 and SOCS3 transcript levels, but not promoter methylation levels in subcutaneous adipose tissues from women with obesity. Moreover, mRNA expression of SOCS1 and SOCS3 in SAT was associated with known obesity indices, insulin resistance, and hs-CRP, suggesting the contribution of SOCS1 and SOCS3 in the pathogenesis of obesity-related metabolic abnormalities. However, further studies are required to establish this concept.
Collapse
Affiliation(s)
- Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fataneh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran Sciences, Tehran, Iran
| | - Seyedeh Zahra Hasanpour
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Pilehvari
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Zabihi-Mahmoudabadi
- Department of Surgery, School of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Meysam Motevasseli
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shanaki
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Filipović M, Flegar D, Aničić S, Šisl D, Kelava T, Kovačić N, Šućur A, Grčević D. Transcriptome profiling of osteoclast subsets associated with arthritis: A pathogenic role of CCR2 hi osteoclast progenitors. Front Immunol 2022; 13:994035. [PMID: 36591261 PMCID: PMC9797520 DOI: 10.3389/fimmu.2022.994035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The existence of different osteoclast progenitor (OCP) subsets has been confirmed by numerous studies. However, pathological inflammation-induced osteoclastogenesis remains incompletely understood. Detailed characterization of OCP subsets may elucidate the pathophysiology of increased osteoclast activity causing periarticular and systemic bone resorption in arthritis. In our study, we rely on previously defined OCP subsets categorized by the level of CCR2 expression as circulatory-like committed CCR2hi OCPs, which are substantially expanded in arthritis, and marrow-resident CCR2lo OCPs of immature phenotype and behavior. Methods In order to perform transcriptome characterization of those subsets in the context of collagen-induced arthritis (CIA), we sorted CCR2hi and CCR2lo periarticular bone marrow OCPs of control and arthritic mice, and performed next-generation RNA sequencing (n=4 for each group) to evaluate the differential gene expression profile using gene set enrichment analysis with further validation. Results A disparity between CCR2hi and CCR2lo subset transcriptomes (863 genes) was detected, with the enrichment of pathways for osteoclast differentiation, chemokine and NOD-like receptor signaling in the CCR2hi OCP subset, and ribosome biogenesis in eukaryotes and ribosome pathways in the CCR2lo OCP subset. The effect of intervention (CIA) within each subset was greater in CCR2hi (92 genes) than in CCR2lo (43 genes) OCPs. Genes associated with the osteoclastogenic pathway (Fcgr1, Socs3), and several genes involved in cell adhesion and migration (F11r, Cd38, Lrg1) identified the CCR2hi subset and distinguish CIA from control group, as validated by qPCR (n=6 for control mice, n=9 for CIA mice). The latter gene set showed a significant positive correlation with arthritis clinical score and frequency of CCR2hi OCPs. Protein-level validation by flow cytometry showed increased proportion of OCPs expressing F11r/CD321, CD38 and Lrg1 in CIA, indicating that they could be used as disease markers. Moreover, osteoclast pathway-identifying genes remained similarly expressed (Fcgr1) or even induced by several fold (Socs3) in preosteoclasts differentiated in vitro from CIA mice compared to pre-cultured levels, suggesting their importance for enhanced osteoclastogenesis of the CCR2hi OCPs in arthritis. Conclusion Our approach detected differentially expressed genes that could identify distinct subset of OCPs associated with arthritis as well as indicate possible therapeutic targets aimed to modulate osteoclast activity.
Collapse
Affiliation(s)
- Maša Filipović
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Darja Flegar
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sara Aničić
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Dino Šisl
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Alan Šućur
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia,*Correspondence: Alan Šućur, ; Danka Grčević,
| | - Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia,Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia,*Correspondence: Alan Šućur, ; Danka Grčević,
| |
Collapse
|
13
|
Rossetti AC, Paladini MS, Brüning CA, Spero V, Cattaneo MG, Racagni G, Papp M, Riva MA, Molteni R. Involvement of the IL-6 Signaling Pathway in the Anti-Anhedonic Effect of the Antidepressant Agomelatine in the Chronic Mild Stress Model of Depression. Int J Mol Sci 2022; 23:ijms232012453. [PMID: 36293308 PMCID: PMC9604470 DOI: 10.3390/ijms232012453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 12/03/2022] Open
Abstract
Neuroinflammation has emerged as an important factor in the molecular underpinnings of major depressive disorder (MDD) pathophysiology and in the mechanism of action of antidepressants. Among the inflammatory mediators dysregulated in depressed patients, interleukin (IL)-6 has recently been proposed to play a crucial role. IL-6 activates a signaling pathway comprising the JAK/STAT proteins and characterized by a specific negative feedback loop exerted by the cytoplasmic protein suppressor of cytokine signalling-3 (SOCS3). On these bases, here, we explored the potential involvement of IL-6 signaling in the ability of the antidepressant drug agomelatine to normalize the anhedonic-like phenotype induced in the rat by chronic stress exposure. To this aim, adult male Wistar rats were subjected to the chronic mild stress (CMS) paradigm and chronically treated with vehicle or agomelatine. The behavioral evaluation was assessed by the sucrose consumption test, whereas molecular analyses were performed in the prefrontal cortex. We found that CMS was able to stimulate IL-6 production and signaling, including SOCS3 gene and protein expression, but the SOCS3-mediated feedback-loop inhibition failed to suppress the IL-6 cascade in stressed animals. Conversely, agomelatine treatment normalized the stress-induced decrease in sucrose consumption and restored the negative modulation of the IL-6 signaling via SOCS3 expression and activity. Our results provide additional information about the pleiotropic mechanisms that contribute to agomelatine’s therapeutic effects.
Collapse
Affiliation(s)
- Andrea C. Rossetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Cesar Augusto Brüning
- Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas, Pelotas 96010-900, RS, Brazil
| | - Vittoria Spero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Marco A. Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
- Correspondence:
| |
Collapse
|
14
|
Li Y, Liu X, Tu R, Hou J, Zhuang G. Mendelian Randomization Analysis of the Association of SOCS3 Methylation with Abdominal Obesity. Nutrients 2022; 14:nu14183824. [PMID: 36145200 PMCID: PMC9503364 DOI: 10.3390/nu14183824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
This study was conducted to evaluate the potential causality association of SOCS3 methylation with abdominal obesity using Mendelian randomization. A case-control study, including 1064 participants, was carried out on Chinese subjects aged 18 to 79. MethylTargetTM was used to detect the methylation level for each CpG site of SOCS3, and SNPscan® was applied to measure the single-nucleotide polymorphism (SNP) genotyping. The logistic regression was used to assess the relationship of SOCS3 methylation level and SNP genotyping with abdominal obesity. Three types of Mendelian randomization methods were implemented to examine the potential causality between SOCS3 methylation and obesity based on the SNP of SOCS3 as instrumental variables. SOCS3 methylation levels were inversely associated with abdominal obesity in five CpG sites (effect estimates ranged from 0.786 (Chr17:76356054) to 0.851 (Chr17:76356084)), and demonstrated positively association in 18 CpG sites (effect estimates ranged from 1.243 (Chr17:76354990) to 1.325 (Chr17:76355061)). The causal relationship between SOCS3 methylation and abdominal obesity was found using the maximum-likelihood method and Mendelian randomization method of penalized inverse variance weighted (MR-IVW), and the β values (95% CI) were 5.342 (0.215, 10.469) and 4.911 (0.259, 9.564), respectively. The causality was found between the SOCS3 methylation level and abdominal obesity in the Chinese population.
Collapse
Affiliation(s)
- Yuqian Li
- Departmentof Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
- Department of Clinical Pharmacology, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Xiaotian Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Zhengzhou University, Zhengzhou 450052, China
| | - Runqi Tu
- Department of Epidemiology and Biostatistics, School of Public Health, Zhengzhou University, Zhengzhou 450052, China
| | - Jian Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Zhengzhou University, Zhengzhou 450052, China
| | - Guihua Zhuang
- Departmentof Epidemiology and Biostatistics, School of Public Health, Xi’an Jiaotong University Health Science Centre, Xi’an 710061, China
- Correspondence: ; Tel.: +86-29-826-551-03
| |
Collapse
|
15
|
Yu C, Fan Y, Zhang Y, Liu L, Guo G. LINC00893 inhibits the progression of prostate cancer through miR-3173-5p/SOCS3/JAK2/STAT3 pathway. Cancer Cell Int 2022; 22:228. [PMID: 35818076 PMCID: PMC9275192 DOI: 10.1186/s12935-022-02637-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 06/17/2022] [Indexed: 11/21/2022] Open
Abstract
Background Prostate cancer (PCa) is one of the most common malignant tumors in the male urinary system. In recent years, the morbidity and mortality of PCa have been increasing due to the limited effects of existing treatment strategies. Long non-coding RNA (lncRNA) LINC00893 was reported to inhibit the proliferation and metastasis of papillary thyroid cancer cells, but its role in PCa has not been reported. This study aims to investigate the role and underlying mechanism of LINC00893 in regulating the progression of PCa cells. Methods We first compared LINC00893 expression levels between PCa tissues and normal prostate tissues through TCGA database. The relative LINC00893 expression levels were further validated in 66 pairs of PCa tissues and para-cancerous normal tissues, as well as in PCa cell lines. Gain-of-function experiment was performed by transfecting PCa cell with LINC00893 expression vector, and CCK (Cell count kit)-8, 5-Ethynyl-2′-deoxyuridine (EdU) incorporation, colony information and transwell assays were conducted to assess the functional phenotypes. Dual-luciferase reporter, RNA-binding protein immunoprecipitation (RIP) and RNA pull-down assays were performed to evaluate the molecular interactions. Results LINC00893 was downregulated in PCa tissues and cell lines, and patients with low expression of LINC00893 were associated with a poorer overall survival rate. LINC00893 overexpression hindered the proliferation, epithelial-mesenchymal transition (EMT) as well as the migratory ability of PCa cells, and suppressed the tumorigenesis of PCa cells in nude mice. We further demonstrated that LINC00893 acted as a sponge for miR-3173-5p and inhibited its activity, which in turn regulated the suppressor of cytokine signaling 3 (SOCS3)/Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling axis. Conclusions Our study demonstrated that LINC00893 suppresses the progression of PCa cells through targeting miR-3173-5p/SOCS3/JAK2/STAT3 axis. Our data uncovers a novel tumor-suppressor role of LINC00893 in PCa, which may serve as a potential strategy for targeted therapy in PCa. Grapical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02637-4.
Collapse
Affiliation(s)
- Chuigong Yu
- Department of Urology, The Third Medical Center, Chinese People's Liberation Army General Hospital, No. 69, Yongding Road, Haidian District, Beijing, 100039, China
| | - Yu Fan
- Department of Urology, The Third Medical Center, Chinese People's Liberation Army General Hospital, No. 69, Yongding Road, Haidian District, Beijing, 100039, China
| | - Yu Zhang
- Department of Urology, The Third Medical Center, Chinese People's Liberation Army General Hospital, No. 69, Yongding Road, Haidian District, Beijing, 100039, China
| | - Lupeng Liu
- Department of Urology, The Third Medical Center, Chinese People's Liberation Army General Hospital, No. 69, Yongding Road, Haidian District, Beijing, 100039, China
| | - Gang Guo
- Department of Urology, The Third Medical Center, Chinese People's Liberation Army General Hospital, No. 69, Yongding Road, Haidian District, Beijing, 100039, China.
| |
Collapse
|
16
|
Xiao Y, Wang Z, Zhao M, Ji W, Xiang C, Li T, Wang R, Yang K, Qian C, Tang X, Xiao H, Zou Y, Liu H. A novel defined risk signature of interferon response genes predicts the prognosis and correlates with immune infiltration in glioblastoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:9481-9504. [PMID: 35942769 DOI: 10.3934/mbe.2022441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Interferons (IFNs) have been implemented as anti-tumor immunity agents in clinical trials of glioma, but only a subset of glioblastoma (GBM) patients profits from it. The predictive role of IFNs stimulated genes in GBM needs further exploration to investigate the clinical role of IFNs. METHODS This study screened 526 GBM patients from three independent cohorts. The transcriptome data with matching clinical information were analyzed using R. Immunohistochemical staining data from the Human Protein Atlas and DNA methylation data from MethSurv were used for validation in protein and methylation level respectively. RESULTS We checked the survival effect of all 491 IFNs response genes, and found 54 genes characterized with significant hazard ratio in overall survival (OS). By protein-protein interaction analysis, 10 hub genes were selected out for subsequent study. And based on the expression of these 10 genes, GBM patients could be divided into two subgroups with significant difference in OS. Furthermore, the least absolute shrinkage and selection operator cox regression model was utilized to construct a multigene risk signature, including STAT3, STAT2 and SOCS3, which could serve as an independent prognostic predictor for GBM. The risk model was validated in two independent GBM cohorts. The GBM patients with high risk scores mainly concentrated in the GBM Mesenchymal subtype. The higher risk group was enriched in hypoxia, angiogenesis, EMT, glycolysis and immune pathways, and had increased Macrophage M2 infiltration and high expression of immune checkpoint CD274 (namely PD-L1). CONCLUSIONS Our findings revealed the three-gene risk model could be an independent prognostic predictor for GBM, and they were crucial participants in immunosuppressive microenvironment of GBM.
Collapse
Affiliation(s)
- Yong Xiao
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhen Wang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Wei Ji
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Chong Xiang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, Changzhou Wujin People's Hospital, Changzhou, China
| | - Taiping Li
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Ran Wang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Kun Yang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chunfa Qian
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xianglong Tang
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yuanjie Zou
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Li L, Zhang J, Peng H, Jiang X, Liu Z, Tian H, Hou S, Xie X, Peng Q, Zhou T. Knockdown of miR-92a suppresses the stemness of colorectal cancer cells via mediating SOCS3. Bioengineered 2022; 13:5613-5624. [PMID: 35184640 PMCID: PMC8974062 DOI: 10.1080/21655979.2021.2022267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
MiRNAs (microRNAs) participate in colorectal cancer (CRC) progression and act as potential biomarkers for CRC prognosis. In this study, we investigated the mechanisms of microRNA-92a (miR-92a) in CRC. Expressions of miR-92a and SOCS3 (Suppressor Of Cytokine Signaling 3) were investigated by qRT-PCR in CRC cell lines and 30 cases of CRC. The self-renewal capacity and proliferation of CRC stem cells were estimated by the sphere formation assay, EdU staining, and Flow cytometry analysis. Moreover, the interplay between miR-92a and SOCS3 in CRC cells was validated by luciferase reporter experiments. MiR-92a was found to be remarkably increased while SOCS3 was significantly downregulated in CRC tissues. Inhibition of miR-92a or SOCS3 attenuated the sphere formation capacity, decreased expressions of stemness-related proteins, and inhibited the proliferation of cancer stem-like cells. Knockdown of SOCS3 reversed the repressive impacts of miR-92a inhibitors on self-renewal and growth of CRC cancer stem cells. This study suggested that miR-92a functions as an oncogene of CRC through mediating the stemness of colorectal cancer cells by directly binding and repressing SOCS3.
Collapse
Affiliation(s)
- Lifa Li
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jingxiao Zhang
- Department of Medica, The Second Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hong Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, the Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xianhong Jiang
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zuoliang Liu
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongpeng Tian
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Songlin Hou
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xingjiang Xie
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiang Peng
- Department of Gastrointestinal Surgery II, Hepatobiliary and Pancreatic Research, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tong Zhou
- Department of Gastrointestinal Surgery II, The Second Affiliated Hospital of North Sichuan Medical College, Sichuan, China
- Department of Gastrointestinal Surgery II, Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| |
Collapse
|
18
|
Wan L, Jia RM, Ji LL, Qin XM, Hu L, Hu F, Han Y, Pan YB, Jiang CY, Liu WT. AMPK-autophagy-mediated inhibition of microRNA-30a-5p alleviates morphine tolerance via SOCS3-dependent neuroinflammation suppression. J Neuroinflammation 2022; 19:25. [PMID: 35093117 PMCID: PMC8800317 DOI: 10.1186/s12974-022-02384-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The development of morphine tolerance is a clinical challenge for managing severe pain. Studies have shown that neuroinflammation is a critical aspect for the development of analgesic tolerance. We found that AMPK-autophagy activation could suppress neuroinflammation and improve morphine tolerance via the upregulation of suppressor of cytokine signaling 3 (SOCS3) by inhibiting the processing and maturation of microRNA-30a-5p. METHODS CD-1 mice were utilized for the tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 was utilized to investigate the mechanism of AMPK-autophagy-mediated posttranscriptional regulation of SOCS3. Proinflammatory cytokines were measured by western blotting and real-time PCR. The levels of SOCS3 and miRNA-processing enzymes were evaluated by western blotting, real-time PCR and immunofluorescence staining. RESULTS Based on experimental verification, miRNA-30a-5p could negatively regulate SOCS3. The AMPK activators AICAR, resveratrol and metformin downregulated miRNA-30a-5p. We found that AMPK activators specifically inhibited the processing and maturation of miRNA-30a-5p in microglia by degrading DICER and AGO2 via autophagy. Furthermore, a miRNA-30a-5p inhibitor significantly improved morphine tolerance via upregulation of SCOS3 in mice. It markedly increased the level of SOCS3 in the spinal cord of mice and subsequently inhibited morphine-induced phosphorylation of NF-κB p65. In addition, a miRNA-30a-5p inhibitor decreased the levels of IL-1β and TNF-α caused by morphine in microglia. CONCLUSION AMPK-autophagy activation suppresses neuroinflammation and improves morphine tolerance via the upregulation of SOCS3 by inhibiting miRNA-30a-5p.
Collapse
Affiliation(s)
- Li Wan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ru-Meng Jia
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Lu-Lu Ji
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xin-Miao Qin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Fan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuan Han
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yin-Bing Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chun-Yi Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
19
|
La Manna S, De Benedictis I, Marasco D. Proteomimetics of Natural Regulators of JAK-STAT Pathway: Novel Therapeutic Perspectives. Front Mol Biosci 2022; 8:792546. [PMID: 35047557 PMCID: PMC8762217 DOI: 10.3389/fmolb.2021.792546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
The JAK-STAT pathway is a crucial cellular signaling cascade, including an intricate network of Protein-protein interactions (PPIs) responsible for its regulation. It mediates the activities of several cytokines, interferons, and growth factors and transduces extracellular signals into transcriptional programs to regulate cell growth and differentiation. It is essential for the development and function of both innate and adaptive immunities, and its aberrant deregulation was highlighted in neuroinflammatory diseases and in crucial mechanisms for tumor cell recognition and tumor-induced immune escape. For its involvement in a multitude of biological processes, it can be considered a valuable target for the development of drugs even if a specific focus on possible side effects associated with its inhibition is required. Herein, we review the possibilities to target JAK-STAT by focusing on its natural inhibitors as the suppressor of cytokine signaling (SOCS) proteins. This protein family is a crucial checkpoint inhibitor in immune homeostasis and a valuable target in immunotherapeutic approaches to cancer and immune deficiency disorders.
Collapse
Affiliation(s)
| | | | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
20
|
Yu Y, Sung SK, Lee CH, Ha M, Kang J, Kwon EJ, Kang JW, Kim Y, Kim GH, Heo HJ, Lee H, Kim TW, Lee Y, Myung K, Oh CK, Kim YH. SOCS3 is Related to Cell Proliferation in Neuronal Tissue: An Integrated Analysis of Bioinformatics and Experiments. Front Genet 2021; 12:743786. [PMID: 34646310 PMCID: PMC8502821 DOI: 10.3389/fgene.2021.743786] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Glioma is the most common primary malignant tumor that occurs in the central nervous system. Gliomas are subdivided according to a combination of microscopic morphological, molecular, and genetic factors. Glioblastoma (GBM) is the most aggressive malignant tumor; however, efficient therapies or specific target molecules for GBM have not been developed. We accessed RNA-seq and clinical data from The Cancer Genome Atlas, the Chinese Glioma Genome Atlas, and the GSE16011 dataset, and identified differentially expressed genes (DEGs) that were common to both GBM and lower-grade glioma (LGG) in three independent cohorts. The biological functions of common DEGs were examined using NetworkAnalyst. To evaluate the prognostic performance of common DEGs, we performed Kaplan-Meier and Cox regression analyses. We investigated the function of SOCS3 in the central nervous system using three GBM cell lines as well as zebrafish embryos. There were 168 upregulated genes and 50 downregulated genes that were commom to both GBM and LGG. Through survival analyses, we found that SOCS3 was the only prognostic gene in all cohorts. Inhibition of SOCS3 using siRNA decreased the proliferation of GBM cell lines. We also found that the zebrafish ortholog, socs3b, was associated with brain development through the regulation of cell proliferation in neuronal tissue. While additional mechanistic studies are necessary, our results suggest that SOCS3 is an important biomarker for glioma and that SOCS3 is related to the proliferation of neuronal tissue.
Collapse
Affiliation(s)
- Yeuni Yu
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Busan, South Korea
| | - Soon Ki Sung
- Department of Neurosurgery, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Chi Hyung Lee
- Department of Neurosurgery, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Mihyang Ha
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan, South Korea
| | - Junho Kang
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Busan, South Korea
| | - Eun Jung Kwon
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan, South Korea
| | - Ji Wan Kang
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan, South Korea
| | - Youngjoo Kim
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan, South Korea
| | - Ga Hyun Kim
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan, South Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Hansong Lee
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Busan, South Korea
| | - Tae Woo Kim
- Department of Orthopaedic Surgery, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, South Korea.,Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, South Korea
| | - Chang-Kyu Oh
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, South Korea.,Department of Anatomy, College of Medicine, Inje University, Busan, South Korea
| | - Yun Hak Kim
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Busan, South Korea.,Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| |
Collapse
|
21
|
Li J, Pan X, Ren Z, Li B, Liu H, Wu C, Dong X, de Vos P, Pan LL, Sun J. Protein arginine methyltransferase 2 (PRMT2) promotes dextran sulfate sodium-induced colitis by inhibiting the SOCS3 promoter via histone H3R8 asymmetric dimethylation. Br J Pharmacol 2021; 179:141-158. [PMID: 34599829 DOI: 10.1111/bph.15695] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE There is emerging evidence for a critical role for epigenetic modifiers in the development of inflammatory bowel disease (IBD). Protein arginine methyltransferase 2 (PRMT2) is responsible for the methylation of arginine residues on histones and targets transcription factors involved in many cellular processes, including gene transcription, mRNA splicing, cell proliferation, and cell differentiation. In this study, the role and underlying mechanisms of PRMT2 in colitis were studied. EXPERIMENTAL APPROACH A mouse dextran sulfate sodium (DSS)-induced experimental colitis model was used to study PRMT2 in colitis. Lentivirus-induced PRMT2 silencing or overexpression in vivo was applied to address the role of PRMT2 in colitis. Detailed western blot and expression analysis were done to understand epigenetic changes induced by PRMT2 in colitis. KEY RESULTS PRMT2 is highly expressed in inflammatory bowel disease patients, in inflamed murine colon and in TNF-α stimulated murine gut epithelial cells. PRMT2 overexpression aggravates, while knockdown alleviates DSS-induced colitis, suggesting that PRMT2 is a pivotal mediator of colitis in mice. Mechanistically, PRMT2 mediates colitis by increasing repressive histone mark H3R8 asymmetric methylation (H3R8me2a) at the promoter region of the suppressor of cytokine signalling 3 promoter (SOCS3). Resultant inhibition of SOCS3 expression and inhibition of SOCS3-mediated degradation of TNF receptor associated factor 5 (TRAF5) via ubiquitination led to elevated TRAF5 expression and TRAF5-mediated downstream NF-κB/MAPK activation. CONCLUSION AND IMPLICATIONS Our study demonstrates that PRMT2 acts as a transcriptional co-activator for proinflammatory genes during colitis. Hence, targeting PRMT2 may provide a novel therapeutic approach for colitis.
Collapse
Affiliation(s)
- Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhengnan Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Binbin Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - He Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xiaoliang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
22
|
Huang B, Chen H, Zheng Y. MiR-103/miR-107 inhibits enterovirus 71 replication and facilitates type I interferon response by regulating SOCS3/STAT3 pathway. Biotechnol Lett 2021; 43:1357-1369. [PMID: 33796959 DOI: 10.1007/s10529-021-03115-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Enterovirus71 (EV71), the major cause of hand, foot, and-mouth disease (HFMD), has increasingly become a public health challenge. Type I interferons (IFNs) can regulate innate and adaptive immune responses to pathogens. MicroRNAs (miRNAs) play regulatory roles in host innate immune responses to viral infections. However, the roles of miR-103 and miR-107 in EV71 infection remain unclear. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to determine the expression of miR-103, miR-107, suppressor of cytokine signaling 3 (SOCS3), VP1, IFN-α, and IFN-β. Virus titers were measured by 50% tissue culture infectious dose (TCID50) assay. Western blot assay was conducted to detect the protein levels of VP1, IFN-α, IFN-β, SOCS3, signal transducer and activator of transcription 3 (STAT3), and phospho-STAT3 (p-STAT3). Immunofluorescence assay was used to detect the protein level of VP1. The concentrations of IFN-α and IFN-β were examined by Enzyme-linked immunosorbent assay (ELISA). The interaction between SOCS3 and miR-103/miR-107 was predicted by starBase and verified by dual-luciferase reporter assay and RNA pull-down assay. RESULTS MiR-103 and miR-107 were downregulated and SOCS3 was upregulated in serum from patients with EV71 and EV71-infected cells. Overexpression of miR-103 and miR-107 repressed EV71 replication by inhibiting EV71 titers and VP1 expression. Moreover, upregulation of miR-103 and miR-107 enhanced EV71-triggered the production of type I IFNs. In addition, miR-103 and miR-107 directly targeted SOCS3, and SOCS3 upregulation reversed the effects of miR-103 and miR-107 on EV71 replication and type I IFN response. Importantly, miR-103 and miR-107 increased STAT3 phosphorylation by targeting SOCS3 after EV71 infection. CONCLUSION MiR-103 and miR-107 suppressed EV71 replication and increased the production of type I IFNs by regulating SOCS3/STAT3 pathway, which might provide a novel strategy for developing effective antiviral therapy.
Collapse
Affiliation(s)
- Baizhi Huang
- Department of Pediatrics, Binhaiwan Central Hospital of Dongguan, Dongguan, China.
- Department of Pediatrics, Binhaiwan Central Hospital of Dongguan, No. 111 Humen Avenue, Humen Town, Dongguan City, 523900, Guangdong Province, China.
| | - Haiping Chen
- Department of Pediatrics, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Yanbing Zheng
- Department of Pediatrics, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| |
Collapse
|
23
|
Xia Y, Lei C, Yang D, Luo H. Identification of key modules and hub genes associated with lung function in idiopathic pulmonary fibrosis. PeerJ 2020; 8:e9848. [PMID: 33194355 PMCID: PMC7485506 DOI: 10.7717/peerj.9848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial lung disease, characterized by a decline in lung function. To date, the pathophysiologic mechanisms associated with lung dysfunction remain unclear, and no effective therapy has been identified to improve lung function. Methods In the present study, we used weighted gene co-expression network analysis (WGCNA) to identify key modules and hub genes associated with lung function in IPF. Three datasets, containing clinical information, were downloaded from Gene Expression Omnibus. WGCNA was performed on the GSE32537 dataset. Differentially expressed gene s (DEGs) between IPF patients and healthy controls were also identified to filter hub genes. The relationship between hub genes and lung function was then validated using the GSE47460 and GSE24206 datasets. Results The red module, containing 267 genes, was positively correlated with the St. George’s Respiratory Questionnaire score (r = 0.37, p < 0.001) and negatively correlated with the percent predicted forced vital capacity (FVC% predicted) (r = − 0.46, p < 0.001) and the percent predicted diffusion capacity of the lung for carbon monoxide (Dlco% predicted) (r = − 0.42, p < 0.001). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis suggested that the genes in the red module were primarily involved in inflammation and immune pathways. Based on Module Membership and Gene Significance, 32 candidate hub genes were selected in the red module to construct a protein-protein interaction network . Based on the identified DEGs and the degree of connectivity in the network, we identified three hub genes, including interleukin 6 (IL6), suppressor of cytokine signaling-3 (SOCS3), and serpin family E member 1 (SERPINE1). In the GSE47460 dataset, Spearman correlation coefficients between Dlco% predicted and expression levels of IL6, SERPINE1, SOCS3 were –0.32, –0.41, and –0.46, respectively. Spearman correlation coefficients between FVC% predicted and expression levels of IL6, SERPINE1, SOCS3 were –0.29, –0.33, and –0.27, respectively. In the GSE24206 dataset, all three hub genes were upregulated in patients with advanced IPF. Conclusion We identified three hub genes that negatively correlated with the lung function of IPF patients. Our results provide insights into the pathogenesis underlying the progressive disruption of lung function, and the identified hub genes may serve as biomarkers and potential therapeutictargets for the treatment of IPF patients.
Collapse
Affiliation(s)
- Yuechong Xia
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, Hunan, China
| | - Cheng Lei
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, Hunan, China
| | - Danhui Yang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, Hunan, China
| | - Hong Luo
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.,Hunan Diagnosis and Treatment Center of Respiratory Disease, Changsha, Hunan, China
| |
Collapse
|
24
|
Effect of Lactobacillus rhamnosus GG on Energy Metabolism, Leptin Resistance, and Gut Microbiota in Mice with Diet-Induced Obesity. Nutrients 2020; 12:nu12092557. [PMID: 32846917 PMCID: PMC7551584 DOI: 10.3390/nu12092557] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is closely associated with various metabolic disorders, including leptin resistance, which is characterized by high circulating leptin levels. Probiotics can decrease circulating leptin levels by alteration of the gut microbiota. Thus, they may have anti-obesogenic effects. In this study, the effects of administration of a probiotic bacterium, Lactobacillus rhamnosus GG (LGG), on gut microbiota and modulation of leptin resistance were evaluated in mice. Male Balb/C mice aged 7 weeks were fed either a normal diet (ND), high-fat diet (HFD), HFD supplemented with low-dose LGG (108 CFU/mouse/day), or HFD supplemented with high-dose LGG (1010 CFU/mouse/day) for 10 weeks. Significantly increased body weight, epididymal fat weight, and decreased leptin responsiveness to exogenous leptin treatment and ratio of villus height to crypt depth were observed in the HFD-fed mice compared to the ND-fed mice. Moreover, a remarkable increase in the proportion of Proteobacteria and ratio of Firmicutes/Bacteroidetes in the fecal microbiota were also observed in the HFD-fed mice. Supplementation of HFD with high-dose LGG restored exogenous leptin responsiveness, increased the ratio of villus height to crypt depth, and decreased the proportion of Proteobacteria in fecal microbiota. These findings suggest that LGG supplementation might alleviate leptin resistance caused by an HFD through the improvement of the digestive health of the host.
Collapse
|
25
|
Yu Q, Yang X, Zhang C, Zhang X, Wang C, Chen L, Liu X, Gu Y, He X, Hu L, Liu WT, Li Y. AMPK activation by ozone therapy inhibits tissue factor-triggered intestinal ischemia and ameliorates chemotherapeutic enteritis. FASEB J 2020; 34:13005-13021. [PMID: 32776374 DOI: 10.1096/fj.201902717rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
Chemotherapeutic enteritis is a major dose-limiting adverse reaction to chemotherapy, with few effective drugs in clinic. Intestinal ischemic injury plays prominent role in chemotherapeutic enteritis clinically. However, mechanism is not clear. In this article, irinotecan (CPT-11) was used to establish chemotherapeutic enteritis mice model. Western blotting, gelatin zymography, immunohistochemistry (IHC), Laser Doppler flowmetry (LDF) were used to detect the pathogenesis of ischemia-hypoxia injury. CPT-11 increased levels of tissue factor (TF) both in the blood and in intestines, and decreased the intestinal blood flow in mice. Interestingly, the elevation of TF in the blood displayed "double-peak," which was consistent with the intestinal mucosal "double-strike" injury trend. Intestinal microthrombus and mixed thrombus formation were detectable in chemotherapeutic enteritis. Furthermore, ozone therapy relieved chemotherapeutic enteritis in mice. Ozone inhibited TF expression induced by CPT-11 via activating AMPK/SOCS3, and effectively ameliorated the intestinal mucosal injury in mice. Moreover, ozone autotransfusion therapy effectively attenuated chemotherapeutic enteritis and the blood hypercoagulability in patients. For the first time, we proposed that TF-induced thrombotic intestinal ischemic injury is a core trigger pathological mechanism of chemotherapeutic enteritis, and provided a new treatment strategy, ozone therapy, to suppress TF expression and treat chemotherapeutic enteritis.
Collapse
Affiliation(s)
- Qingqing Yu
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, P.R. China.,Department of Oncology, Jining No.1 People's Hospital, Jining, P.R. China
| | - Xing Yang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Chen Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Xiaotao Zhang
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, P.R. China.,Department of Radiation Oncology, Qingdao Central Hospital, Qingdao, P.R. China
| | - Chaoyu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Lu Chen
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Xiaolin Liu
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, P.R. China
| | - Yufeng Gu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Xueming He
- Center for Clinical Research and Translational Medicine, The Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, P.R. China.,Institute of Translational Medicine, Nanjing Medical University, Nanjing, P.R. China
| | - Yan Li
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, P.R. China.,Department of Oncology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, P.R. China
| |
Collapse
|
26
|
Ahmed SAH, Ansari SA, Mensah-Brown EPK, Emerald BS. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus. Clin Epigenetics 2020; 12:104. [PMID: 32653024 PMCID: PMC7353744 DOI: 10.1186/s13148-020-00896-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic condition characterised by β cell dysfunction and persistent hyperglycaemia. The disorder can be due to the absence of adequate pancreatic insulin production or a weak cellular response to insulin signalling. Among the three types of DM, namely, type 1 DM (T1DM), type 2 DM (T2DM), and gestational DM (GDM); T2DM accounts for almost 90% of diabetes cases worldwide. Epigenetic traits are stably heritable phenotypes that result from certain changes that affect gene function without altering the gene sequence. While epigenetic traits are considered reversible modifications, they can be inherited mitotically and meiotically. In addition, epigenetic traits can randomly arise in response to environmental factors or certain genetic mutations or lesions, such as those affecting the enzymes that catalyse the epigenetic modification. In this review, we focus on the role of DNA methylation, a type of epigenetic modification, in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Sanabil Ali Hassan Ahmed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Eric P K Mensah-Brown
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
27
|
Wang S, Lv J, Meng S, Tang J, Nie L. Recent Advances in Nanotheranostics for Treat-to-Target of Rheumatoid Arthritis. Adv Healthc Mater 2020; 9:e1901541. [PMID: 32031759 DOI: 10.1002/adhm.201901541] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/31/2019] [Indexed: 12/16/2022]
Abstract
Early diagnosis, standardized treatment, and regular monitoring are the clinical treatment principle of rheumatoid arthritis (RA). The overarching principles and recommendations of treat-to-target (T2T) in RA advocate remission as the optimum aim, especially for patients with very early disease who are initiating therapy with anti-RA medications. However, traditional anti-RA drugs cannot selectively target the inflammatory areas and may cause serious side effects due to its short biological half-life and poor bioavailability. These limitations have significantly driven the research and application of nanomaterial-based drugs in theranostics of RA. Nanomedicines have appropriate sizes and easily modified surfaces which can enhance their biological compatibility and prolong circulation time of drug-loading systems in vivo. Traditional T2T regimens cannot evaluate the efficacy of drugs in real time, while clinical drug nanosizing can realize the integration of diagnosis and treatment of RA. This review bridges clinically proposed T2T concepts and nanomedicine in an integrated system for RA early-stage diagnosis and treatment. The most advanced progress in various nanodrug delivery systems for theranostics of RA is summarized, establishing a clear path and a new perspective for further optimization of T2T. Finally, the key facing challenges are discussed and prospects are addressed.
Collapse
Affiliation(s)
- Shasha Wang
- Hunan Key Laboratory of Biomedical Nanomaterials and DevicesHunan University of Technology Zhuzhou 412007 P. R. China
| | - Jing Lv
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen University Xiamen 361102 P. R. China
| | - Shanshan Meng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen University Xiamen 361102 P. R. China
| | - Jianxin Tang
- Hunan Key Laboratory of Biomedical Nanomaterials and DevicesHunan University of Technology Zhuzhou 412007 P. R. China
| | - Liming Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen University Xiamen 361102 P. R. China
| |
Collapse
|
28
|
Induction of SOCS Expression by EV71 Infection Promotes EV71 Replication. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2430640. [PMID: 32149091 PMCID: PMC7054758 DOI: 10.1155/2020/2430640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/03/2020] [Indexed: 12/30/2022]
Abstract
Enterovirus 71 (EV71) is the causative pathogen of hand, foot, and mouth disease (HFMD). However, no effective antiviral therapy is currently available. Some viruses could escape the host's innate immunity by upregulating suppressor of cytokine signaling (SOCS) proteins. Until now, whether EV71 evades the host immune system by regulating the expression of SOCS proteins remains unknown. In this study, we found that EV71 infection promoted SOCS expression at both mRNA and protein levels in vitro and in vivo. Consistently, the infectivity of EV71 was decreased significantly in the SOCS3 or SOCS1 knockdown cells, suggesting that SOCS1 and especially SOCS3 are crucial for EV71 infection. Further investigation showed that SOCS3 promoted virus infection by inhibiting interferon-induced STAT3 phosphorylation. SOCS1 and SOCS3 mRNA expressions were independent on virus-induced type I interferon expression but were blocked by the inhibitor of NF-κB. Therefore, EV71 infection stimulates the expression of SOCS proteins in an interferon-independent way and negatively regulates the JAK/STAT signaling pathway, thus escaping host immunity. All these results may add new information to the mechanism of EV71 in fighting against type I interferon responses.
Collapse
|
29
|
Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, Ma H, Wei D, Sun S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020; 80:106210. [PMID: 31972425 DOI: 10.1016/j.intimp.2020.106210] [Citation(s) in RCA: 466] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 02/09/2023]
Abstract
The JAK/STAT signaling pathway is an universally expressed intracellular signal transduction pathway and involved in many crucial biological processes, including cell proliferation, differentiation, apoptosis, and immune regulation. It provides a direct mechanism for extracellular factors-regulated gene expression. Current researches on this pathway have been focusing on the inflammatory and neoplastic diseases and related drug. The mechanism of JAK/STAT signaling is relatively simple. However, the biological consequences of the pathway are complicated due to its crosstalk with other signaling pathways. In addition, there is increasing evidence indicates that the persistent activation of JAK/STAT signaling pathway is closely related to many immune and inflammatory diseases, yet the specific mechanism remains unclear. Therefore, it is necessary to study the detailed mechanisms of JAK/STAT signaling in disease formation to provide critical reference for clinical treatments of the diseases. In this review, we focus on the structure of JAKs and STATs, the JAK/STAT signaling pathway and its negative regulators, the associated diseases, and the JAK inhibitors for the clinical therapy.
Collapse
Affiliation(s)
- Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Xiaoyun Xu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Youzhi Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuegang Zhou
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Najing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Najing 211200, China
| | - Donghua Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shiqin Sun
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
30
|
Fruit and vegetable intake modifies the associations between suppressor of cytokine signaling 3 genetic variants and type 2 diabetes. Eur J Nutr 2020; 59:3441-3449. [PMID: 31927672 DOI: 10.1007/s00394-020-02178-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Type 2 diabetes is a complex disease determined by variable genes and environmental factors. The study was designed to investigate the effect of interactions of four polymorphisms of suppressor of cytokine signaling 3 (SOCS3) with fruit and vegetable (F&V) intake on type 2 diabetes in a rural population of China. METHODS A total of 4411 participants from the rural areas of Henan, China were included in the study. Multivariate logistic regression and restricted cubic splines were used to estimate the associations between polymorphisms and risk allele score of SOCS3 and type 2 diabetes in different groups. Haplotype analysis was conducted to examine the effects of linkage inheritance at these four loci on type 2 diabetes. RESULTS Three of the four polymorphisms showed significant associations with type 2 diabetes in the less F&V intake group after adjusting the covariates, the odds ratios (ORs) and corresponding 95% confidence intervals (95% CIs) were 1.24 (1.08-1.41) for rs4969168, 1.16 (1.02-1.32) for rs9892622, and 1.21 (1.06-1.39) for rs9914220. No significant association was detected in the more F&V intake group. The obvious dose-response relationship between the risk allele score and type 2 diabetes was also noted only in the less F&V intake group. CONCLUSIONS Variants of SOCS3 gene were associated with type 2 diabetes and the associations could be modified by the F&V intake.
Collapse
|
31
|
Nozaki Y, Ri J, Sakai K, Niki K, Kinoshita K, Funauchi M, Matsumura I. Inhibition of the IL-18 Receptor Signaling Pathway Ameliorates Disease in a Murine Model of Rheumatoid Arthritis. Cells 2019; 9:E11. [PMID: 31861496 PMCID: PMC7017073 DOI: 10.3390/cells9010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Interleukin (IL)-18 expression in synovial tissue correlates with the severity of joint inflammation and the levels of pro-inflammatory cytokines. However, the role of the IL-18/IL-18 receptor-alpha (Rα) signaling pathway in autoimmune arthritis is unknown. Wild-type (WT) and IL-18Rα knockout (KO) mice were immunized with bovine type II collagen before the onset of arthritis induced by lipopolysaccharide injection. Disease activity was evaluated by semiquantitative scoring and histologic assessment. Serum inflammatory cytokine and anticollagen antibody levels were quantified by an enzyme-linked immunosorbent assay. Joint cytokine and matrix metalloproteinases-3 levels were determined by a quantitative polymerase chain reaction. Splenic suppressors of cytokine signaling (SOCS) were determined by Western blot analysis as indices of systemic immunoresponse. IL-18Rα KO mice showed lower arthritis and histological scores in bone erosion and synovitis due to reductions in the infiltration of CD4+ T cells and F4/80+ cells and decreased serum IL-6, -18, TNF, and IFN-γ levels. The mRNA expression and protein levels of SOCS3 were significantly increased in the IL-18Rα KO mice. By an up-regulation of SOCS, pro-inflammatory cytokines were decreased through the IL-18/IL-18Rα signaling pathway. These results suggest that inhibitors of the IL-18/IL-18Rα signaling pathway could become new therapeutic agents for rheumatoid arthritis.
Collapse
Affiliation(s)
- Yuji Nozaki
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine; Osaka-sayama, Osaka 589-8511, Japan; (J.R.); (K.S.); (K.N.); (K.K.); (M.F.); (I.M.)
| | | | | | | | | | | | | |
Collapse
|
32
|
Fazeli E, Piltan S, Sadeghi H, Gholami M, Azizi-Tabesh G, Yassaee F, Mirfakhraie R. Ectopic expression of CYP24A1 circular RNA hsa_circ_0060927 in uterine leiomyomas. J Clin Lab Anal 2019; 34:e23114. [PMID: 31746073 PMCID: PMC7171336 DOI: 10.1002/jcla.23114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/03/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Background As a novel class of non‐coding RNAs, the role of circular RNAs (circRNAs) in tumor biogenesis and progression has been proved in a number of human tumors; however, up to now, the relation between circRNAs and uterine leiomyomas (ULM) remains unclear. Methods In this study, we have estimated the expression level of CYP24A1 hsa_circ_0060927 in uterine leiomyoma and adjacent tissues considering the mediator complex subunit 12 gene (MED12) mutation profile by quantitative real‐time polymerase chain reaction (qRT‐PCRs). Results Using Sanger sequencing method, somatic mutations in the MED12 exon 2 were detected in 14 (35.90%) ULM samples, including 10 (71.43%) missense mutations and 4 (28.57%) in‐frame deletions. Our results revealed that hsa_circ_0060927 was ectopically expressed in 33.33% of ULM tissues; although, this expression was independent of the MED12 mutation profile in the ULM samples. Conclusions Present results provide primary evidence for the role of circular RNAs in the leiomyoma development; however, further studies are essential to confirm the importance of these molecules as potential biomarkers for diagnosis and/or prognosis in ULM.
Collapse
Affiliation(s)
- Elnaz Fazeli
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Piltan
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Sadeghi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Gholami
- Department of Biochemistry and Genetics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ghasem Azizi-Tabesh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fakhrolmolook Yassaee
- Department of Obstetrics and Gynecology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Zheng Y, Hou X, Yang S. Lidocaine Potentiates SOCS3 to Attenuate Inflammation in Microglia and Suppress Neuropathic Pain. Cell Mol Neurobiol 2019; 39:1081-1092. [PMID: 31209627 DOI: 10.1007/s10571-019-00703-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 06/08/2019] [Indexed: 01/10/2023]
Abstract
Lidocaine is one of the typical local anesthetics that are frequently used in the peripheral nerve blocks and pain management. Emerging evidence have shown that lidocaine may exert anti-inflammatory effect involving neuropathic pain. However, the effect and underlying mechanism of lidocaine in suppressing neuroinflammation in neuropathic pain are incompletely revealed. In this study, effects of lidocaine on the suppressors of cytokine-signaling protein 3 (SOCS3) in microglia are investigated in chronic constriction injury (CCI) rat model and lipopolysaccharide (LPS)-stimulated BV-2 cells. It was shown that intrathecal injection of lidocaine substantially alleviated CCI-induced neuropathic pain, as reflected by the decreased thermal latency and mechanical threshold. Lidocaine reduced the CCI-evoked spinal injury and cell apoptosis. CCI induced an significant increase of IBA1+ microglia accompanied by the increase of inflammatory cytokines IL-6 and IL-1β, which were suppressed after lidocaine administration. SOCS3 expression in IBA1+ microglia was notably upregulated in response to lidocaine injection, which presented in a similar pattern in LPS-activated BV-2 cells. Furthermore, lidocaine upregulated SOCS3 expression dependent of pCREB, and CREB silencing greatly discounted this effect. The intrathecal injection of lentiviral vectors LV-SOCS3 efficiently alleviated CCI-evoked neuropathic pain and reduced spinal IBA1+ microglia. SOCS3 overexpression contributed to the inhibition of neuroinflammation by decreasing the expression and activation of p38 MAPK and NF-κB stimulated by LPS. Collectively, lidocaine promoted the SOCS3 expression in microglia, in turn leading to suppression of IBA1+ microglia accumulation and p38 MAPK and NF-κB, which may expand our understanding on lidocaine in suppressing neuroinflammation and neuropathic pain.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Anesthesiology, China-Japan Union Hospital, Jilin University, Changchun, 130033, Jilin, China
| | - Xuhui Hou
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, No. 126 Sendai Street, Changchun, 130033, Jilin, China
| | - Songbai Yang
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, No. 126 Sendai Street, Changchun, 130033, Jilin, China.
| |
Collapse
|
34
|
Korovina I, Neuwirth A, Sprott D, Troullinaki M, Poitz DM, Deussen A, Klotzsche-von Ameln A. Myeloid SOCS3 Deficiency Regulates Angiogenesis via Enhanced Apoptotic Endothelial Cell Engulfment. J Innate Immun 2019; 12:248-256. [PMID: 31574508 DOI: 10.1159/000502645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/12/2019] [Indexed: 01/05/2023] Open
Abstract
Mononuclear phagocytes, such as macrophages and microglia, are key regulators of organ homeostasis including vascularization processes. Here, we investigated the role of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells as a regulator of mononuclear phagocyte function and their interaction with endothelial cells in the context of sprouting angiogenesis. As compared to SOCS3-sufficient counterparts, SOCS3-deficient microglia and macrophages displayed an increased phagocytic activity toward primary apoptotic endothelial cells, which was associated with an enhanced expression of the opsonin growth arrest-specific 6 (Gas6), a major prophagocytic molecule. Furthermore, we found that myeloid SOCS3 deficiency significantly reduced angiogenesis in an ex vivo mouse aortic ring assay, which could be reversed by the inhibition of the Gas6 receptor Mer. Together, SOCS3 in myeloid cells regulates the Gas6/Mer-dependent phagocytosis of endothelial cells, and thereby angiogenesis-related processes. Our findings provide novel insights into the complex crosstalk between mononuclear phagocytes and endothelial cells, and may therefore provide a new platform for the development of new antiangiogenic therapies.
Collapse
Affiliation(s)
- Irina Korovina
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,OncoRay, National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ales Neuwirth
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - David Sprott
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Maria Troullinaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anne Klotzsche-von Ameln
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany, .,Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany,
| |
Collapse
|
35
|
Pentagalloylglucose Inhibits the Replication of Rabies Virus via Mediation of the miR-455/SOCS3/STAT3/IL-6 Pathway. J Virol 2019; 93:JVI.00539-19. [PMID: 31243136 DOI: 10.1128/jvi.00539-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Our previous study showed that pentagalloylglucose (PGG), a naturally occurring hydrolyzable phenolic tannin, possesses significant anti-rabies virus (RABV) activity. In BHK-21 cells, RABV induced the overactivation of signal transducer and activator of transcription 3 (STAT3) by suppressing the expression of suppressor of cytokine signaling 3 (SOCS3). Inhibition of STAT3 by niclosamide, small interfering RNA, or exogenous expression of SOCS3 all significantly suppressed the replication of RABV. Additionally, RABV-induced upregulation of microRNA 455-5p (miR-455-5p) downregulated SOCS3 by directly binding to the 3' untranslated region (UTR) of SOCS3. Importantly, PGG effectively reversed the expression of miR-455-5p and its following SOCS3/STAT3 signaling pathway. Finally, activated STAT3 elicited the expression of interleukin-6 (IL-6), thereby contributing to RABV-associated encephalomyelitis; however, PGG restored the level of IL-6 in vitro and in vivo in a SOCS3/STAT3-dependent manner. Altogether, these data identify a new miR-455-5p/SOCS3/STAT3 signaling pathway that contributes to viral replication and IL-6 production in RABV-infected cells, with PGG exerting its antiviral effect by inhibiting the production of miR-455-5p and the activation of STAT3.IMPORTANCE Rabies virus causes lethal encephalitis in mammals and poses a serious public health threat in many parts of the world. Numerous strategies have been explored to combat rabies; however, their efficacy has always been unsatisfactory. We previously reported a new drug, PGG, which possesses a potent inhibitory activity on RABV replication. Herein, we describe the underlying mechanisms by which PGG exerts its anti-RABV activity. Our results show that RABV induces overactivation of STAT3 in BHK-21 cells, which facilitates viral replication. Importantly, PGG effectively inhibits the activity of STAT3 by disrupting the expression of miR-455-5p and increases the level of SOCS3 by directly targeting the 3' UTR of SOCS3. Furthermore, the downregulated STAT3 inhibits the production of IL-6, thereby contributing to a reduction in the inflammatory response in vivo Our study indicates that PGG effectively inhibits the replication of RABV by the miR-455-5p/SOCS3/STAT3/IL-6-dependent pathway.
Collapse
|
36
|
Yakass MB, Franco D, Quaye O. Suppressors of Cytokine Signaling and Protein Inhibitors of Activated Signal Transducer and Activator of Transcriptions As Therapeutic Targets in Flavivirus Infections. J Interferon Cytokine Res 2019; 40:1-18. [PMID: 31436502 DOI: 10.1089/jir.2019.0097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Flaviviruses cause significant human diseases putting more than 400 million people at risk annually worldwide. Because of migration and improved transportation, these viruses can be found on all continents (except Antarctica). Although a majority of the viruses are endemic in the tropics, a few [West Nile virus (WNV) and tick-borne encephalitis virus (TBEV)] have shown endemicity in Europe and North America. Currently, there are vaccines for the Yellow fever virus, Japanese encephalitis virus, and TBEV, but there is no effective vaccine and/or therapy against all other flaviviruses. Although there are intensive efforts to develop vaccines for Zika viruses, dengue viruses, and WNVs, there is the need for alternative or parallel antiviral therapeutic approaches. Suppressors of cytokine signaling (SOCS) and protein inhibitors of activated signal transducer and activator of transcription (STATs; PIAS), both regulatory proteins of the Janus kinase/STAT signaling pathway, have been explored as therapeutic targets in herpes simplex and vaccinia viruses, as well as in cancer therapy. In this review, we briefly describe the function of SOCS and PIAS and their therapeutic potential in flaviviral infections. [Figure: see text].
Collapse
Affiliation(s)
- Michael Bright Yakass
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | | | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| |
Collapse
|
37
|
Wang B, Wangkahart E, Secombes CJ, Wang T. Insights into the Evolution of the Suppressors of Cytokine Signaling (SOCS) Gene Family in Vertebrates. Mol Biol Evol 2019; 36:393-411. [PMID: 30521052 PMCID: PMC6368001 DOI: 10.1093/molbev/msy230] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The SOCS family are key negative regulators of cytokine and growth factor signaling. Typically, 8-17 SOCS genes are present in vertebrate species with eight known in mammals, classified as type I (SOCS4-7) and type II (CISH and SOCS1-3) SOCS. It was believed that the type II SOCS were expanded through the two rounds of whole genome duplication (1R and 2R WGDs) from a single CISH/SOCS1-3 precursor. Previously, 12 genes were identified in rainbow trout but here we report 15 additional loci are present, and confirm 26 of the genes are expressed, giving rainbow trout the largest SOCS gene repertoire identified to date. The discovery of the additional SOCS genes in trout has led to a novel model of SOCS family evolution, whereby the vertebrate SOCS gene family was derived from CISH/SOCS2, SOCS1/SOCS3, SOCS4/5, SOCS6, and SOCS7 ancestors likely present before the two WGD events. It is also apparent that teleost SOCS2b, SOCS4, and SOCS5b molecules are not true orthologues of mammalian SOCS2, SOCS4, and SOCS5, respectively. The rate of SOCS gene structural changes increased from 2R vertebrates, to 4R rainbow trout, and the genes with structural changes show large differences and low correlation coefficient of expression levels relative to their paralogues, suggesting a role of structural changes in expression and functional diversification. This study has important impacts in the functional prediction and understanding of the SOCS gene family in different vertebrates, and provides a framework for determining how many SOCS genes could be expected in a particular vertebrate species/lineage.
Collapse
Affiliation(s)
- Bei Wang
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, College of Fishery, Guangdong Ocean University, Zhanjiang, P.R. China.,Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Eakapol Wangkahart
- Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Research Unit of Excellence for Tropical Fisheries and Technology, Division of Fisheries, Department of Agricultural Technology, Faculty of Technology, Mahasarakham University, Khamriang Sub-District, Kantarawichai, Mahasarakham, Thailand
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Tiehui Wang
- Scottish Fish Immunology Research Centre, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
38
|
Convery O, Gargan S, Kickham M, Schroder M, O'Farrelly C, Stevenson NJ. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3. FASEB J 2019; 33:8732-8744. [PMID: 31163989 DOI: 10.1096/fj.201800629rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viruses use a spectrum of immune evasion strategies that enable infection and replication. The acute phase of hepatitis C virus (HCV) infection is characterized by nonspecific and often mild clinical symptoms, suggesting an immunosuppressive mechanism that, unless symptomatic liver disease presents, allows the virus to remain largely undetected. We previously reported that HCV induced the regulatory protein suppressor of cytokine signaling (SOCS)3, which inhibited TNF-α-mediated inflammatory responses. However, the mechanism by which HCV up-regulates SOCS3 remains unknown. Here we show that the HCV protein, p7, enhances both SOCS3 mRNA and protein expression. A p7 inhibitor reduced SOCS3 induction, indicating that p7's ion channel activity was required for optimal up-regulation of SOCS3. Short hairpin RNA and chemical inhibition revealed that both the Janus kinase-signal transducer and activator of transcription (JAK-STAT) and MAPK pathways were required for p7-mediated induction of SOCS3. HCV-p7 expression suppressed TNF-α-mediated IκB-α degradation and subsequent NF-κB promoter activity, revealing a new and functional, anti-inflammatory effect of p7. Together, these findings identify a molecular mechanism by which HCV-p7 induces SOCS3 through STAT3 and ERK activation and demonstrate that p7 suppresses proinflammatory responses to TNF-α, possibly explaining the lack of inflammatory symptoms observed during early HCV infection.-Convery, O., Gargan, S., Kickham, M., Schroder, M., O'Farrelly, C., Stevenson, N. J. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3.
Collapse
Affiliation(s)
- Orla Convery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
39
|
Cevey ÁC, Penas FN, Alba Soto CD, Mirkin GA, Goren NB. IL-10/STAT3/SOCS3 Axis Is Involved in the Anti-inflammatory Effect of Benznidazole. Front Immunol 2019; 10:1267. [PMID: 31214200 PMCID: PMC6558013 DOI: 10.3389/fimmu.2019.01267] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Anti-parasitic treatment for Chagas disease mainly relies on benznidazole, which is virtually the only drug available in the market. Besides its anti-parasitic effects, benznidazole has anti-inflammatory properties. In this work we studied the mechanisms involved in the latter, demonstrating the participation of the IL-10/STAT3/SOCS3 pathway. To achieve this goal, the anti-inflammatory properties of benznidazole were studied using an in vitro model of cardiomyocyte primary culture stimulated with LPS. LPS increased both SOCS3 expression and STAT3 phosphorylation. The addition of benznidazole increased their expression even further. Specific inhibition of STAT3 precluded this effect, suggesting a role for STAT3 in the increase of SOCS3 expression induced by benznidazole. To assess the participation of SOCS3 in the anti-inflammatory effect of benznidazole, we accomplished specific knockdown of SOCS3 with siRNA. Silencing of SOCS3 in cardiomyocytes precluded the inhibitory effects of benznidazole on TNF-α, IL-6, iNOS expression and NO release. Moreover, in the absence of SOCS3, benznidazole could neither prevent IKK phosphorylation nor IκBα degradation, supporting the notion that SOCS3 is required for the benznidazole-mediated inhibition of the NF-κB pathway. Previously, we demonstrated that IL-10 increases the expression of SOCS3 in cultured cardiomyocytes. Here, we found that benznidazole shows a trend to increased IL-10 expression. To evaluate whether benznidazole increased SOCS3 in an IL-10-dependent manner, cardiomyocytes from IL-10 knockout mice were pre-treated with benznidazole and stimulated with LPS. Benznidazole neither inhibited NO release nor avoid IKK phosphorylation or IκBα degradation, showing that IL-10 is required for benznidazole-mediated inhibition of NF-κB. Moreover, exogenous addition of IL-10 to IL-10 knockout cardiomyocytes restored the inhibitory effect of benznidazole on NO release. The results reported herein show, for the first time, that the IL-10/STAT3/SOCS3 axis is involved in the anti-inflammatory effects of benznidazole. These findings may add up to new therapeutic strategies for chronic Chagas disease given its inflammatory nature.
Collapse
Affiliation(s)
- Ágata C Cevey
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico N Penas
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Catalina D Alba Soto
- Facultad de Medicina, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gerardo A Mirkin
- Facultad de Medicina, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora B Goren
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
40
|
Liu K, Wu Z, Chu J, Yang L, Wang N. Promoter methylation and expression of SOCS3 affect the clinical outcome of pediatric acute lymphoblastic leukemia by JAK/STAT pathway. Biomed Pharmacother 2019; 115:108913. [PMID: 31054507 DOI: 10.1016/j.biopha.2019.108913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Suppressor of cytokine signaling 3 (SOCS3) has been characterized as one of the most crucial negative regulator in the JAK2/STAT3 signaling pathway. However, there are few studies on the relationship between SOCS3 and pediatric acute lymphoblastic leukemia (ALL). This study analyzes the influence of SOCS3 expression on the risk and the progression of pediatric ALL and the underlying mechanism. The levels of SOCS3, p-JAK2, p-STAT3, SOCS3 methylation, CD4+CD25+CD127lowTreg were detected by PCR, laser confocal microscopy, western blot, bisulfite sequencing and flow cytometry at different progression of ALL. We found that the SOCS3 expression level at initial diagnosis (DG) of ALL patients was significantly lower than that of healthy controls (HC), while the expression of SOCS3 methylation was opposite. The expression of SOCS3 and SOCS3 methylation were returned to normal in the complete remission (CR) stage, and there were no difference between resistance, relapse and initial diagnosis. The expression of SOCS3 decreased and weakened the inhibition of pSTAT3 expression in DG, resistance and relapse groups. The levels of Treg cells in ALL children were significantly higher than those in the HC children. There was a positive correlation between the expression level of STAT3 and the expression level of Treg cells in children with ALL, while that was negatively correlated with the expression levels of Treg cells. Compared with high-level of SOCS3, the low-level of SOCS3 patients had more high risk factors, as higher WBC counts, LDH level and much more poor prognostic genes. SOCS3 methylation leads to low-expression of SOCS3, which can lead to continuous activation of JAK/STAT3 and increased expression of Treg cells, which in turn affects the anti-tumor immunological effect of the body. Taken together, our data show that monitoring the level of SOCS3 can contribute to the understanding of the state of illness and evaluate the risk of progression of ALL.
Collapse
Affiliation(s)
- Kangkang Liu
- Pediatrics, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhengyu Wu
- Pediatrics, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jinhua Chu
- Pediatrics, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Linhai Yang
- Pediatrics, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ningling Wang
- Pediatrics, the Second Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
41
|
Yin Y, Guo R, Shao Y, Ge M, Miao C, Cao L, Yang Y, Hu L. Pretreatment with resveratrol ameliorate trigeminal neuralgia by suppressing matrix metalloproteinase-9/2 in trigeminal ganglion. Int Immunopharmacol 2019; 72:339-347. [PMID: 31009895 DOI: 10.1016/j.intimp.2019.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/29/2019] [Accepted: 04/05/2019] [Indexed: 01/05/2023]
Abstract
Trigeminal neuralgia (TN) is a common type of neuropathic pain whereas the underlying pathogenesis has not been completely elucidated. Recent study suggests that the development of neuroinflammation is responsible for generating and sustaining neuropathic pain. The purpose of our study was to investigate the protective effect of intervening the inflammation in early stages of pain and explore its potential mechanism. MMP-9 and MMP-2 are vital proinflammatory participants and accumulating evidence indicates that they are involved in the early development of neuropathic pain. In this study, we found that MMP-9/2 showed different temporal up regulation in trigeminal ganglion (TG) significantly after chronic constriction injury (CCI) surgery. However, the activation of MMP-9/2 were suppressed by the pretreatment with resveratrol, which delayed and attenuated CCI-induced mechanical allodynia simultaneously. Besides, the expression of proinflammatory cytokines like IL-1β and TNF-α as well as the excessive neuronal activity induced by CCI were suppressed by resveratrol. Moreover, we believed that the inhibition of MMP-9/2 activation and pain sensitization may be related to the TLR-4/NF-κB signaling pathway, which might be negatively regulated by the induction of SOCS3. In conclusion, pretreatment with resveratrol could be an effective approach to alleviate trigeminal neuralgia in early stages via a powerful inhibition on the activation of MMP-9/2 in TG.
Collapse
Affiliation(s)
- Yuling Yin
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Rong Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yu Shao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Mixue Ge
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Chen Miao
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Ling Cao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yanjing Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| | - Liang Hu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
42
|
Curto P, Riley SP, Simões I, Martinez JJ. Macrophages Infected by a Pathogen and a Non-pathogen Spotted Fever Group Rickettsia Reveal Differential Reprogramming Signatures Early in Infection. Front Cell Infect Microbiol 2019; 9:97. [PMID: 31024862 PMCID: PMC6467950 DOI: 10.3389/fcimb.2019.00097] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Despite their high degree of genomic similarity, different spotted fever group (SFG) Rickettsia are often associated with very different clinical presentations. For example, Rickettsia conorii causes Mediterranean spotted fever, a life-threatening disease for humans, whereas Rickettsia montanensis is associated with limited or no pathogenicity to humans. However, the molecular basis responsible for the different pathogenicity attributes are still not understood. Although killing microbes is a critical function of macrophages, the ability to survive and/or proliferate within phagocytic cells seems to be a phenotypic feature of several intracellular pathogens. We have previously shown that R. conorii and R. montanensis exhibit different intracellular fates within macrophage-like cells. By evaluating early macrophage responses upon insult with each of these rickettsial species, herein we demonstrate that infection with R. conorii results in a profound reprogramming of host gene expression profiles. Transcriptional programs generated upon infection with this pathogenic bacteria point toward a sophisticated ability to evade innate immune signals, by modulating the expression of several anti-inflammatory molecules. Moreover, R. conorii induce the expression of several pro-survival genes, which may result in the ability to prolong host cell survival, thus protecting its replicative niche. Remarkably, R. conorii-infection promoted a robust modulation of different transcription factors, suggesting that an early manipulation of the host gene expression machinery may be key to R. conorii proliferation in THP-1 macrophages. This work provides new insights into the early molecular processes hijacked by a pathogenic SFG Rickettsia to establish a replicative niche in macrophages, opening several avenues of research in host-rickettsiae interactions.
Collapse
Affiliation(s)
- Pedro Curto
- Ph.D. Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Sean P. Riley
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Isaura Simões
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Juan J. Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
43
|
Pérez-Baos S, Gratal P, Barrasa JI, Lamuedra A, Sánchez-Pernaute O, Herrero-Beaumont G, Largo R. Inhibition of pSTAT1 by tofacitinib accounts for the early improvement of experimental chronic synovitis. JOURNAL OF INFLAMMATION-LONDON 2019; 16:2. [PMID: 30728752 PMCID: PMC6352431 DOI: 10.1186/s12950-019-0206-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/20/2019] [Indexed: 01/24/2023]
Abstract
Background In order to gain insight into the early effects drawn by JAK inhibitors on intra-joint JAK/STAT-dependent signaling, we sought synovial activation of STATs and their end-products, along with their modification with tofacitinib (TOFA), at flare-up in antigen induced arthritis (AIA). New Zealand rabbits were randomly assigned to four groups –healthy controls, AIA, TOFA-treated AIA, or TOFA-treated controls–. AIA was induced with 4 weekly intra-articular ovalbumin injections in sensitized animals. TOFA (10 mg·kg− 1·day− 1) was administered for the last 2 weeks. Animals were euthanized 24 h after the last injection. Results AIA animals showed high-grade synovitis, which was partially improved by TOFA. No effects of the treatment were found on serum C-reactive protein or on the synovial macrophage infiltration at this stage. Synovial MMP-1,-3 and -13 expression levels in treated AIA rabbits were found to drop to those of controls, while a downregulation of IL6, IFNγ and TNF was evident in treated versus untreated AIA rabbits. Concurrently, a reduction in pSTAT1 and SOCS1, but not in pSTAT3, SOCS3 or active NFκB-p65, was noted with TOFA. Conclusions Studying the mechanism of action of immunomodulatory drugs represents a major challenge in vivo, since drug-dependent decreases in inflammation very likely mask direct effects on disease mechanisms. This study design allowed us to prevent any confounding effect resulting from reductions in the overall inflammatory status, hence assessing the true pharmacological actions of TOFA in a very severe synovitis. Our findings point to pSTAT1 and MMPs as early molecular readouts of response to this JAK inhibitor. Electronic supplementary material The online version of this article (10.1186/s12950-019-0206-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandra Pérez-Baos
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| | - Paula Gratal
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| | - Juan I Barrasa
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain.,2Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
| | - Ana Lamuedra
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| | - Olga Sánchez-Pernaute
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| | - Gabriel Herrero-Beaumont
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| | - Raquel Largo
- 1Bone and Joint Research Unit, Rheumatology Department, IIS-Fundación Jiménez Díaz UAM, Avenida Reyes Católicos, 2. 28040, Madrid, Spain
| |
Collapse
|
44
|
SOCS4 expressed by recombinant HSV protects against cytokine storm in a mouse model. Oncol Rep 2018; 41:1509-1520. [PMID: 30569160 PMCID: PMC6365695 DOI: 10.3892/or.2018.6935] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/30/2018] [Indexed: 12/26/2022] Open
Abstract
Oncolytic viruses are genetically engineered viruses designed for the treatment of solid tumors, and are often coupled with the antitumor immunity of the host. The challenge of using oncolytic herpes simplex virus (oHSV) as an efficacious oncolytic agent is the potential host tissue damage caused by the production of a range of cytokines following intratumoral oHSV injection. An HSV-suppressor of cytokine signaling 4 (SOCS4) recombinant virus was created to investigate whether it inhibits cytokine storm. Recombinant HSV-SOCS4 and HSV-1(F) were used to infect mice, and levels of several representative cytokines, including monocyte chemoattractant protein-1, interleukin (IL)-1β, tumor necrosis factor-α, IL-6 and interferon γ, in serum and bronchoalveolar lavage fluid (BALF) of infected mice were determined, and immune cells in BALF and spleen were enumerated. Lung damage, virus titers in the lung, body weight and survival rates of infected mice were also determined and compared between the two groups. The cytokine concentration of HSV-SOCS4-infected mice was significantly decreased compared with that of HSV-1(F)-infected mice in BALF and serum, and a smaller number of cluster of differentiation (CD)11b+ cells of BALF, and CD8+CD62L+ T cells and CD4+CD62L+ T cells of the spleen were also identified in HSV-SOCS4-infected mice. HSV-SOCS4-infected mice exhibited slight lung damage, a decrease in body weight loss and a 100% survival rate. The results of the present study indicated that SOCS4 protein may be a useful regulator to inhibit cytokine overproduction, and that HSV-SOCS4 may provide a possible solution to control cytokine storm and its consequences following induction by oncolytic virus treatment.
Collapse
|
45
|
Pulmonary phagocyte-derived NPY controls the pathology of severe influenza virus infection. Nat Microbiol 2018; 4:258-268. [PMID: 30455472 DOI: 10.1038/s41564-018-0289-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/10/2018] [Indexed: 01/21/2023]
Abstract
Crosstalk between the autonomic nervous system and the immune system by means of the sympathetic and parasympathetic pathways is a critical process in host defence. Activation of the sympathetic nervous system results in the release of catecholamines as well as neuropeptide Y (NPY). Here, we investigated whether phagocytes are capable of the de novo production of NPY, as has been described for catecholamines. We show that the synthesis of NPY and its Y1 receptor (Y1R) is increased in phagocytes in lungs following severe influenza virus infection. The genetic deletion of Npy or Y1r specifically in phagocytes greatly improves the pathology of severe influenza virus infection, which is characterized by excessive virus replication and pulmonary inflammation. Mechanistically, it is the induction of suppressor of cytokine signalling 3 (SOCS3) via NPY-Y1R activation that is responsible for impaired antiviral response and promoting pro-inflammatory cytokine production, thereby enhancing the pathology of influenza virus infection. Thus, direct regulation of the NPY-Y1R-SOCS3 pathway on phagocytes may act as a fine-tuner of an innate immune response to virus infection, which could be a therapeutic target for lethal influenza virus infection.
Collapse
|
46
|
Malemud CJ. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2018; 10:117-127. [PMID: 29942363 PMCID: PMC6009092 DOI: 10.1177/1759720x18776224] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/20/2018] [Indexed: 12/30/2022] Open
Abstract
Proinflammatory cytokine activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signal transduction pathway is a critical event in the pathogenesis and progression of rheumatoid arthritis. Under normal conditions, JAK/STAT signaling reflects the influence of negative regulators of JAK/STAT, exemplified by the suppressor of cytokine signaling and protein inhibitor of activated STAT. However, in rheumatoid arthritis (RA) both of these regulators are dysfunctional. Thus, continuous activation of JAK/STAT signaling in RA synovial joints results in the elevated level of matrix metalloproteinase gene expression, increased frequency of apoptotic chondrocytes and most prominently 'apoptosis resistance' in the inflamed synovial tissue. Tofacitinib, a JAK small molecule inhibitor, with selectivity for JAK2/JAK3 was approved by the United States Food and Drug Administration (US FDA) for the therapy of RA. Importantly, tofacitinib has demonstrated significant clinical efficacy for RA in the post-US FDA-approval surveillance period. Of note, the success of tofacitinib has spurred the development of JAK1, JAK2 and other JAK3-selective small molecule inhibitors, some of which have also entered the clinical setting, whereas other JAK inhibitors are currently being evaluated in RA clinical trials.
Collapse
|
47
|
Sequera C, Manzano S, Guerrero C, Porras A. How Rap and its GEFs control liver physiology and cancer development. C3G alterations in human hepatocarcinoma. Hepat Oncol 2018; 5:HEP05. [PMID: 30302196 PMCID: PMC6168044 DOI: 10.2217/hep-2017-0026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/20/2018] [Indexed: 02/08/2023] Open
Abstract
Rap proteins regulate liver physiopathology. For example, Rap2B promotes hepatocarcinoma (HCC) growth, while Rap1 might play a dual role. The RapGEF, Epac1, activates Rap upon cAMP binding, regulating metabolism, survival, and liver regeneration. A liver specific Epac2 isoform lacking cAMP-binding domain also activates Rap1, promoting fibrosis in alcoholic liver disease. C3G (RapGEF1) is also present in the liver, but mainly as shorter isoforms. Its function in the liver remains unknown. Information from different public genetic databases revealed that C3G mRNA levels increase in HCC, although they decrease in metastatic stages. In addition, several mutations in RapGEF1 gene are present, associated with a reduced patient survival. Based on this, C3G might represent a new HCC diagnostic and prognostic marker, and a therapeutic target.
Collapse
Affiliation(s)
- Celia Sequera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Sara Manzano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer, USAL-CSIC, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, USAL-CSIC, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
48
|
Sun ZL, Jiang XF, Cheng YC, Liu YF, Yang K, Zhu SL, Kong XB, Tu Y, Bian KF, Liu ZL, Chen XY. Exendin-4 inhibits high-altitude cerebral edema by protecting against neurobiological dysfunction. Neural Regen Res 2018; 13:653-663. [PMID: 29722317 PMCID: PMC5950675 DOI: 10.4103/1673-5374.230291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The anti-inflammatory and antioxidant effects of exendin-4 (Ex-4) have been reported previously. However, whether (Ex-4) has anti-inflammatory and antioxidant effects on high-altitude cerebral edema (HACE) remains poorly understood. In this study, two rat models of HACE were established by placing rats in a hypoxic environment with a simulated altitude of either 6000- or 7000-m above sea level (MASL) for 72 hours. An altitude of 7000 MASL with 72-hours of hypoxia was found to be the optimized experimental paradigm for establishing HACE models. Then, in rats where a model of HACE was established by introducing them to a 7000 MASL environment with 72-hours of hypoxia treatment, 2, 10 and, 100 μg of Ex-4 was intraperitoneally administrated. The open field test and tail suspension test were used to test animal behavior. Routine methods were used to detect change in inflammatory cells. Hematoxylin-eosin staining was performed to determine pathological changes to brain tissue. Wet/dry weight ratios were used to measure brain water content. Evans blue leakage was used to determine blood-brain barrier integrity. Enzyme-linked immunosorbent assay (ELISA) was performed to measure markers of inflammation and oxidative stress including superoxide dismutase, glutathione, and malonaldehyde values, as well as interleukin-6, tumor necrosis factor-alpha, cyclic adenosine monophosphate levels in the brain tissue. Western blot analysis was performed to determine the levels of occludin, ZO-1, SOCS-3, vascular endothelial growth factor, EPAC1, nuclear factor-kappa B, and aquaporin-4. Our results demonstrate that Ex-4 preconditioning decreased brain water content, inhibited inflammation and oxidative stress, alleviated brain tissue injury, maintain blood-brain barrier integrity, and effectively improved motor function in rat models of HACE. These findings suggest that Ex-4 exhibits therapeutic potential in the treatment of HACE.
Collapse
Affiliation(s)
- Zhong-Lei Sun
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin; Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xian-Feng Jiang
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin; Tianjin Medical University, Tianjin, China
| | - Yuan-Chi Cheng
- Central Hospital of Fengxian District of Shanghai, Shanghai, China
| | - Ying-Fu Liu
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Kai Yang
- The No. 2 Hospital of Nanjing, Nanjing, Jiangsu Province, China
| | | | - Xian-Bin Kong
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Tu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Ke-Feng Bian
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Zhen-Lin Liu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Xu-Yi Chen
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| |
Collapse
|
49
|
Heppler LN, Frank DA. Targeting Oncogenic Transcription Factors: Therapeutic Implications of Endogenous STAT Inhibitors. Trends Cancer 2017; 3:816-827. [PMID: 29198438 DOI: 10.1016/j.trecan.2017.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023]
Abstract
Misregulation of transcription factors, including signal transducer and activator of transcription (STAT) proteins, leads to inappropriate gene expression patterns that can promote tumor initiation and progression. Under physiologic conditions, STAT signaling is stimulus dependent and tightly regulated by endogenous inhibitors, namely, suppressor of cytokine signaling (SOCS) proteins, phosphatases, and protein inhibitor of activated STAT (PIAS) proteins. However, in tumorigenesis, STAT proteins become constitutively active and promote the expression of progrowth and prosurvival genes. Although STAT activation has been widely implicated in cancer, therapeutic STAT inhibitors are still largely absent from the clinic. This review dissects the mechanisms of action of two families of endogenous STAT inhibitors, the SOCS and PIAS families, to potentially inform the development of novel therapeutic inhibitors.
Collapse
Affiliation(s)
- Lisa N Heppler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Zhang Y, Tao GJ, Hu L, Qu J, Han Y, Zhang G, Qian Y, Jiang CY, Liu WT. Lidocaine alleviates morphine tolerance via AMPK-SOCS3-dependent neuroinflammation suppression in the spinal cord. J Neuroinflammation 2017; 14:211. [PMID: 29096659 PMCID: PMC5667445 DOI: 10.1186/s12974-017-0983-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/18/2017] [Indexed: 12/29/2022] Open
Abstract
Background Morphine tolerance is a clinical challenge, and its pathogenesis is closely related to the neuroinflammation mediated by Toll-like receptor 4 (TLR4). In Chinese pain clinic, lidocaine is combined with morphine to treat chronic pain. We found that lidocaine sufficiently inhibited neuroinflammation induced by morphine and improved analgesic tolerance on the basis of non-affecting pain threshold. Methods CD-1 mice were utilized for tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 was utilized to investigate the mechanism of lidocaine. Neuroinflammation-related cytokines were measured by western blotting and real-time PCR. The level of suppressor of cytokine signaling 3 (SOCS3) and adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK)-related signaling pathway was evaluated by western blotting, real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence staining. Results Lidocaine potentiated an anti-nociceptive effect of morphine and attenuated the chronic analgesic tolerance. Lidocaine suppressed morphine-induced activation of microglia and downregulated inflammatory cytokines, interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α) via upregulating SOCS3 by activating AMPK. Lidocaine enhanced AMPK phosphorylation in a calcium-dependent protein kinase kinase β (CaMKKβ)-dependent manner. Furthermore, lidocaine decreased the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and inhibited the nuclear factor-κB (NF-κB) in accordance with the inhibitory effects to TLR4. Conclusions Lidocaine as a prevalent local anesthetic suppresses morphine tolerance efficiently. AMPK-dependent upregulation of SOCS3 by lidocaine plays a crucial role in the improvement of analgesic tolerance. Electronic supplementary material The online version of this article (10.1186/s12974-017-0983-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Zhang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Research Division of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu, 211100, China
| | - Gao-Jian Tao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Department of Pain, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Liang Hu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jie Qu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yuan Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Guangqin Zhang
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu, 211100, China
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chun-Yi Jiang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Wen-Tao Liu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Department of Pharmacy, Sir Run Run Shaw Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|