1
|
Dozio E, Tassistro E, Orlando A, Giussani M, Beba G, Patti I, Lieti G, Antolini L, Vianello E, Corsi Romanelli MM, Parati G, Genovesi S. The soluble receptor for advanced glycation end products is independently associated with systolic blood pressure values and hypertension in children. Nutr Metab Cardiovasc Dis 2025; 35:103862. [PMID: 39934046 DOI: 10.1016/j.numecd.2025.103862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND AND AIM The advanced glycation end products-receptor for advanced glycation end products (AGE-RAGE) axis is a pro-inflammatory pathway promoting endothelial dysfunction and vascular remodelling. The soluble RAGE form (sRAGE), by blocking circulating AGE, protects against AGE-induced detrimental effects. We investigated the role of sRAGE as a marker of high blood pressure and hypertension risk in children. METHODS AND RESULTS sRAGE was quantified in 284 children/adolescents (mean age (SD) 11.1 (2.5); 52.1 % male) referred for high-normal blood pressure (systolic and/or diastolic values ≥ 90th, but both <95th percentile) or hypertension (systolic and/or diastolic blood pressure ≥95th percentile) and/or other cardiovascular risk factors (excess weight, dyslipidaemia and insulin resistance). In 22.2 % of the sample, systolic and/or diastolic blood pressure values were above the 90th percentile. The prevalence of excess weight (overweight/obesity), central obesity (waist-to-height-ratio >50%), and insulin resistance (HOMA-index ≥90th percentile) was high (82.7 %, 70.8 %, and 70.5 %, respectively). Few children had altered LDL cholesterol, triglyceride, and HDL cholesterol values (15.7 %, 15.4 %, and 13.6 %, respectively). The lowest sRAGE tertile was associated with the highest risk of having hypertension (p = 0.028), obesity (p < 0.001), central obesity (p = 0.007), and insulin resistance (p < 0.001). sRAGE levels were inversely associated with systolic blood pressure (p < 0.01) and BMI (p = 0.022) z-scores and waist-to-height-ratio (p = 0.001). sRAGE values were inversely associated with the presence of hypertension (p = 0.036) and obesity (p = 0.038). CONCLUSIONS The independent relationship between sRAGE, systolic blood pressure, and hypertension in children suggests that the AGE-RAGE axis may be altered early in life, and that sRAGE could be a compelling marker for pediatric cardiovascular risk stratification.
Collapse
Affiliation(s)
- Elena Dozio
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy; Experimental Laboratory for Research on Organ Damage Biomarkers, Istituto Auxologico, IRCCS, Italiano, Milan, Italy
| | - Elena Tassistro
- Biostatistics and Clinical Epidemiology, Fondazione San Gerardo dei Tintori, IRCCS, Monza, Italy; Bicocca Center of Bioinformatics, Biostatistics and Bioimaging (B4 centre), School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Antonina Orlando
- Department of Cardiology, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Marco Giussani
- Department of Cardiology, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Greta Beba
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Ilenia Patti
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Giulia Lieti
- UO Nefrologia e Dialisi, ASST-Rhodense, Garbagnate Milanese, Milan, Italy
| | - Laura Antolini
- Bicocca Center of Bioinformatics, Biostatistics and Bioimaging (B4 centre), School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Elena Vianello
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy; Experimental Laboratory for Research on Organ Damage Biomarkers, Istituto Auxologico, IRCCS, Italiano, Milan, Italy
| | - Massimiliano M Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy; Department of Clinical and Experimental Pathology, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Gianfranco Parati
- Department of Cardiology, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy; School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Simonetta Genovesi
- Department of Cardiology, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy; School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy.
| |
Collapse
|
2
|
Kim HJ, Han CW, Jeong MS, Kwon TJ, Choi JY, Jang SB. Cryo-EM structure of HMGB1-RAGE complex and its inhibitory effect on lung cancer. Biomed Pharmacother 2025; 187:118088. [PMID: 40306174 DOI: 10.1016/j.biopha.2025.118088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
Mitochondrial dysfunction and mitophagy are closely linked with human diseases such as neurodegenerative diseases, metabolic diseases, and cancer. High-mobility group box 1 (HMGB1) has been shown to mediate a wide range of pathological responses by binding with the receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs). Extracellular HMGB1 and its ligand RAGE stimulate the growth, metastasis, invasiveness, and treatment resistance of different cancer cells. Through extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, HMGB1 and RAGE lead to the phosphorylation of Drp1-S616 and Drp1-mediated mitochondrial fission, which consequently causes autophagy. Although the structure of the RAGE and HMGB1 complex is not clearly known, the complex has emerged as a potential therapeutic target. In the present study, the structure of the RAGE and HMGB1 complex was determined at a resolution of 5.19 Å using cryogenic electron microscopy. The structure revealed that the residues P66, G70, P71, S74, and R77 in RAGE and E145, K146, E153, and E156 in HMGB1 were the sites of interaction between the two proteins. Additionally, an HMGB1 peptide (151 LKEKYEK 157) was synthesized based on the RAGE-HMGB1 complex. We investigated the inhibitory function of the HMGB1 peptide and demonstrated that it inhibits tumor growth, metastasis, and invasion by binding to the RAGE protein in lung cancers. The HMGB1 peptide significantly suppressed mitochondrial dysfunction and the initiation of autophagy. Furthermore, the HMGB1 peptide dramatically reduced cell viability, migration, and mitophagy in the colorectal and pancreatic cancer cell lines HCT-116 and AsPC-1, respectively.
Collapse
Affiliation(s)
- Hyeon Jin Kim
- Insitute of Systems Biology, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Republic of Korea.
| | - Chang Woo Han
- Insitute of Systems Biology, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Mi Suk Jeong
- Insitute of Systems Biology, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Republic of Korea.
| | - Tae-Jun Kwon
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80, Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Jun Young Choi
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80, Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Insitute of Systems Biology, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Republic of Korea.
| |
Collapse
|
3
|
Leech T, Kelsell D, Blaydon D, Woodland P, Peiris M. A Role for High Mobility Group Box 1 (HMGB1) Release in the Pathogenesis of Gastroesophageal Reflux Disease. Neurogastroenterol Motil 2025:e70083. [PMID: 40391501 DOI: 10.1111/nmo.70083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/25/2025] [Accepted: 04/21/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Many gastroesophageal reflux disease (GORD) patients have heartburn symptoms despite PPI treatment, potentially via esophageal mucosa exposure to weakly acidic bile salts, leading to oxidative stress. High mobility group box 1 (HMGB1) is a nuclear protein secreted during oxidative stress that binds to receptors including TLR2, TLR4, and RAGE, inducing inflammatory signaling. We describe the release and potential downstream effects of HMGB1 in GORD. METHODS Esophageal biopsies were obtained from healthy controls (HC), functional heartburn (FH), non-erosive reflux disease (NERD), and erosive reflux disease (ERD) patients. Biopsies were analyzed by RNA-sequencing, and the expression of TLR2, TLR4, and RAGE was assessed with immunohistochemistry. NE-1 cells were challenged with pH 5 media and/or 500 μM deoxycholate (DCA), with/without 2-h 5 or 50 μM curcumin pre-treatment. HMGB1 translocation was measured with immunofluorescence and release with ELISA. RESULTS HMGB1, alongside 12 additional oxidative stress-associated genes, is over-expressed in NERD and ERD patients compared to HC (adjusted p < 0.05). Weakly acidic bile salt (pH 5 + DCA) stimulated HMGB1 translocation (p < 0.0001) and release from NE-1 cells (p < 0.001), but ameliorated with curcumin pre-treatment. ERD biopsies had increased DAPI+TLR4+ cells (p < 0.05) and TLR4 fluorescence intensity (p < 0.05), compared to HC. RAGE was expressed on CD45+ cells, and the number of RAGE+CD45+ cells was higher in ERD compared to HC (p < 0.05). CONCLUSIONS Release of HMGB1 from esophageal epithelial cells by weakly acidic bile salts, inhibited by curcumin, and increased expression of TLR4 and RAGE in ERD mucosa suggest this pathway has a role in GORD. Modulating HMGB1 activity may be a strategy for treating recurrent symptoms.
Collapse
Affiliation(s)
- Tom Leech
- Wingate Institute of Neurogastroenterology, Blizard Institute, the Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David Kelsell
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, the Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Diana Blaydon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, the Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Philip Woodland
- Wingate Institute of Neurogastroenterology, Blizard Institute, the Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Madusha Peiris
- Wingate Institute of Neurogastroenterology, Blizard Institute, the Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
4
|
Flores-Estrada J, Cano-Martínez A, Ibarra-Lara L, Jiménez A, Palacios-Reyes C, García LJP, Ortiz-López MG, Rodríguez-Peña ON, Hernández-Portilla LB. Spinach Extract Reduces Kidney Damage in Diabetic Rats by Impairing the AGEs/RAGE Axis. Int J Mol Sci 2025; 26:4730. [PMID: 40429870 PMCID: PMC12111706 DOI: 10.3390/ijms26104730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/02/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The interaction between advanced glycation end products (AGEs) and their RAGE receptor (AGEs/RAGE axis) triggers several signaling pathways that lead to the development of diabetic nephropathy (DN). One of the most studied AGEs is Nε-(1-Carboxymethyl)-L-lysine (CML). Spinacia oleracea is an edible plant with beneficial health properties, but its effect on the AGE/RAGE axis in kidney damage is unknown. OBJECTIVE We aimed to investigate the functional role of spinach methanolic extract (SME) on kidney damage in diabetic rats associated with the CML/RAGE axis. METHODS Forty adult male Wistar rats were used in this study and divided into four groups: control rats (CTRL), SME-administered CTRL (400 mg/kg; SME), streptozotocin-induced diabetic nephropathy rats (STZ), and SME-treated STZ (STZ-SME); treatments were administered daily. After 12 weeks, serum AGEs, creatinine in urine, and lipid peroxidation in kidneys were measured. The distribution and expression levels of inflammatory and fibrotic mediators and RAGE signaling were evaluated through immunohistochemistry (NOX4, CML, RAGE, nuclear NF-κB, TNF-α, IL-1β, TGF-β1, SMAD2/3, CTGF, and a-SMA) and immunolocalization of CML/RAGE. RESULTS Glycoside flavonoid derivatives, such as patuletin and spinacetin, were primarily identified in the extract. Kidneys from the STZ group showed altered morphology, dead cells in the proximal tubules, and increased oxidative stress markers; notably, these effects were improved by SME treatment (STZ-SME). The STZ-SME group showed a lower staining intensity for CML and RAGE, which was associated with a decrease in the expression of inflammatory and fibrotic factors compared with the STZ group. In all groups, the distribution of these markers varied among proximal tubule, glomerular, and interstitial cells. CONCLUSIONS SME treatment may help to prevent or delay kidney damage in diabetic rats by regulating inflammatory and fibrotic processes associated with the AGEs/RAGE pathway, a mechanism involved in the development of nephropathy.
Collapse
Affiliation(s)
- Javier Flores-Estrada
- División de Investigación, Hospital Juárez de México, México City 07760, Mexico; (A.J.); (L.J.P.G.); (M.G.O.-L.)
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico;
| | - Luz Ibarra-Lara
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico;
| | - Adriana Jiménez
- División de Investigación, Hospital Juárez de México, México City 07760, Mexico; (A.J.); (L.J.P.G.); (M.G.O.-L.)
| | - Carmen Palacios-Reyes
- Departamento de Ciencias Médicas, División de Ciencias de la Salud, Universidad de Guanajuato, Campus León, Guanajuato 37000, Mexico;
| | - Luis J. Pinto García
- División de Investigación, Hospital Juárez de México, México City 07760, Mexico; (A.J.); (L.J.P.G.); (M.G.O.-L.)
| | - María G. Ortiz-López
- División de Investigación, Hospital Juárez de México, México City 07760, Mexico; (A.J.); (L.J.P.G.); (M.G.O.-L.)
| | - Olga Nelly Rodríguez-Peña
- Laboratorio de Biogeoquímica, Unidad de Biología, Tecnología y Prototipos (UBIPRO), Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla 54090, Mexico;
| | - Luis Barbo Hernández-Portilla
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Tlalnepantla 54090, Mexico;
| |
Collapse
|
5
|
Feng A, Liang Y, Fu P, Dong Y, Black SM, Wang T. Endotoxin-induced m6A RNA methylation landscape in lung endothelial cells: role of METTL3 in regulating inflammation and injury during acute lung injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167907. [PMID: 40379220 DOI: 10.1016/j.bbadis.2025.167907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Acute Lung Injury (ALI) involves diffuse alveolar damage, neutrophil infiltration, and pulmonary edema, with unacceptable mortality. Bacterial lipopolysaccharide (LPS) activates inflammatory pathways in ALI, which are then regulated by transcriptional and post-transcriptional pathways to affect gene expression. RNA methylation, N6-methyladenosine, is the main m6A mRNA modification that controls the expression of various genes in different environments. There are very few facts about LPS's effect on m6A RNA methylation. This study will explore the m6A RNA methylation landscape in lung endothelial cells (ECs) to understand its role in lung inflammation. In this study, lung endothelial cells were treated with LPS, and the dynamics of mRNA m6A methylation were examined through m6A-methylated RNA sequencing. RNA abundance was measured with RNA-seq, and global protein expression and m6A-binding proteins were identified using mass spectrometry (MS). Following LPS treatment, global m6A methylation levels increased along with the upregulation and nuclear translocation of METTL3 protein, while demethylase activity remained unchanged. METTL3 drove LPS-induced m6A methylation and endothelial injury, as shown by selective METTL3 siRNA and the inhibitor STM2457. MeRIP-seq analyses revealed increased m6A sites near the 5' UTR in LPS-treated cells, with m6A methylation correlating positively with gene expression. The metabolic and apoptosis pathways were shown to be more enriched in different types of methylated exons. METTL3-mediated m6A methylation targeted inflammatory genes, enhancing protein expression in chemokine signaling and MAPK pathways. STM2457 effectively mitigated LPS- or CLP-induced experimental ALI. According to this paper, LPS-mediated m6A RNA methylation is described in terms of genomic structure. Modulation of m6A methylation exerts influence over LPS-mediated endothelial gene expression and the ensuing inflammatory response.
Collapse
Affiliation(s)
- Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
6
|
Settem RP, Sharma A. Oral bacterium contributes to periodontal inflammation by forming advanced glycation end products. Infect Immun 2025; 93:e0056024. [PMID: 40172539 PMCID: PMC12070732 DOI: 10.1128/iai.00560-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
The oral bacterium Tannerella forsythia is associated with periodontitis, an inflammatory disease affecting tooth-supporting tissues. The bacterium produces a dicarbonyl compound, methylglyoxal (MGO), whose levels correlate with the severity of periodontitis. MGO can induce inflammation directly or via the generation of glycation products called advanced glycation end products (AGEs). T. forsythia-produced MGO has been shown to cause tissue collagen glycation, which in turn can induce pro-inflammatory cytokine secretion in monocytes via receptor for advanced glycation end product (RAGE) receptor activation. The current study investigated the impact of T. forsythia-secreted MGO on human gingival fibroblasts and endothelial cells. For assessing the in vivo impact of T. forsythia-secreted MGO, we employed an oral gavage-induced mouse model of periodontitis utilizing the wild-type and MGO-deficient strains of T. forsythia. Our results showed that the apoptotic activity was enhanced, and cell migration was reduced in fibroblasts exposed to collagen treated with the T. forsythia wild-type culture supernatant. Moreover, monocyte binding, reactive oxygen species production, and inflammatory cytokine secretion were increased in fibroblasts, and neutrophil transendothelial migration was enhanced in response to the T. forsythia wild type-treated collagen. In vivo, increased AGE accumulation in gingival tissues with increased alveolar bone loss was observed in wild-type T. forsythia as compared to the MGO-deficient strain-infected mice. These data demonstrated that T. forsythia-secreted MGO contributes to periodontal tissue destruction by mitigating gingival fibroblast-mediated tissue healing and promoting endothelial cell dysfunction. These findings provide a basis for targeting the T. forsythia-associated AGE-RAGE axis in alleviating periodontitis.
Collapse
Affiliation(s)
- Rajendra P. Settem
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, USA
| | - Ashu Sharma
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
7
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [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: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
8
|
Zhong Y, Zhong L, Zhou Y, Liao Y, Deng J. Dynamic neutrophil-to-lymphocyte-platelet ratio trajectories predict 30-day and 1-year mortality in sepsis: a retrospective cohort study based on MIMIC-IV 2.2. BMC Infect Dis 2025; 25:594. [PMID: 40275138 PMCID: PMC12023530 DOI: 10.1186/s12879-025-10987-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND This study investigated the relationship between the neutrophil-to-lymphocyte ratio (N/LPR) and its variability ratio (N/LPRR) with 30-day and 1-year mortality outcomes. METHODS A total of 7,443 patients from the MIMIC-IV 2.2 database were included, with 1,765 having multiple N/LPR measurements. Mortality at 1 year and within 30 days served as the primary endpoints. Patients were stratified into four groups according to baseline N/LPRR quartiles. Receiver operating characteristic (ROC) curves assessed the predictive value of N/LPR and N/LPRR for mortality. Kaplan-Meier analysis estimated the risk of mortality events, while restricted cubic spline (RCS) analysis explored the non-linear associations between N/LPR, N/LPRR, and mortality. Cox proportional hazards regression identified the relationship between N/LPRR and all-cause mortality. RESULTS A total of 792 cases of 1-year mortality (44.9%) were recorded, with 437 deaths (24.8%) occurring within 30 days. ROC analysis revealed that N/LPRR outperformed N/LPR in predicting adverse outcomes. Higher N/LPR and N/LPRR were associated with increased mortality rates. RCS analysis indicated significant non-linear relationships between N/LPR, N/LPRR, and mortality risk (both p-values for nonlinearity < 0.001). Subgroup analyses confirmed the robustness of these findings. CONCLUSION In conclusion, elevated N/LPR and N/LPRR are linked to 30-day and 1-year mortality in patients with sepsis. N/LPRR, with its heightened sensitivity, offers clinicians valuable prognostic information on sepsis severity and progression.
Collapse
Affiliation(s)
- Yuting Zhong
- Department of Anesthesiology, Meizhou People's Hospital, 63 Huangtang Road, Meijiang District, Guangdong, Meizhou, China
| | - Liping Zhong
- Department of Anesthesiology, Meizhou People's Hospital, 63 Huangtang Road, Meijiang District, Guangdong, Meizhou, China
| | - Yuanjun Zhou
- Department of Anesthesiology, Meizhou People's Hospital, 63 Huangtang Road, Meijiang District, Guangdong, Meizhou, China
| | - Yilin Liao
- Department of Anesthesiology, Meizhou People's Hospital, 63 Huangtang Road, Meijiang District, Guangdong, Meizhou, China
| | - Jingdan Deng
- Department of Anesthesiology, Meizhou People's Hospital, 63 Huangtang Road, Meijiang District, Guangdong, Meizhou, China.
| |
Collapse
|
9
|
Radhakrishna U, Radhakrishnan R, Uppala LV, Trivedi TS, Prajapati J, Rawal RM, Muvvala SB, Bahado-Singh RO, Sadhasivam S. Prenatal opioid exposure alters pain perception and increases long-term health risks in infants with neonatal opioid withdrawal syndrome. FRONTIERS IN PAIN RESEARCH 2025; 6:1497801. [PMID: 40313396 PMCID: PMC12043715 DOI: 10.3389/fpain.2025.1497801] [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: 01/12/2025] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Background Opioids are often prescribed for pain relief, yet they pose risks such as addiction, dependence, and overdose. Pregnant women have unique vulnerabilities to opioids and infants born to opioid-exposed mothers could develop neonatal opioid withdrawal syndrome (NOWS). The study of opioid-induced epigenetic changes in chronic pain is in its early stages. This study aimed to identify epigenetic changes in genes associated with chronic pain resulting from maternal opioid exposure during pregnancy. Methods We analyzed DNA methylation of chronic pain-related genes in 96 placental tissues using Illumina Infinium Methylation EPIC BeadChips. These samples comprised 32 from mothers with infants prenatally exposed to opioids who needed pharmacologic NOWS management (+Opioids/+NOWS), 32 from mothers with prenatally opioid-exposed infants not needing NOWS pharmacologic treatment (+Opioids/-NOWS), and 32 from unexposed control subjects (-Opioids/-NOWS). Results The study identified significant methylation changes at 111 CpG sites in pain-related genes among opioid-exposed infants, with 54 CpGs hypomethylated and 57 hypermethylated. These genes play a crucial role in various biological processes, including telomere length regulation (NOS3, ESR1, ESR2, MAPK3); inflammation (TNF, MAPK3, IL1B, IL23R); glucose metabolism (EIF2AK3, CACNA1H, NOTCH3, GJA1); ion channel function (CACNA1C, CACNA1H, CLIC4, KCNQ5); autophagy (CTSS, ULK1, ULK4, ATG5); oxidative stress (NGF, NRG1, OPRM1, ATP1A2); aging (GRIA1, NGFR, PRLR, EIF4E); cytokine activity (TRPV4, RUNX1, CXCL8, IL18R1); and the risk of suicide (ADORA2A, ANKK1, GABRG2, IGSF9B). These epigenetic changes may influence 48 signaling pathways-including cAMP, MAPK, GnRH secretion, estrogen signaling, morphine addiction, circadian rhythms, and insulin secretion-profoundly affecting pain and inflammation-related processes. Conclusion The identified methylation alterations may shed light on pain, neurodevelopmental changes, and other biological mechanisms in opioid-exposed infants and mothers with OUD, offering insights into NOWS and maternal-infant health. These findings may also pave the way for targeted interventions and improved pain management, highlighting the potential for integrated care strategies to address the interconnected health of mothers and infants.
Collapse
Affiliation(s)
- Uppala Radhakrishna
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lavanya V. Uppala
- Department of Pharmacology & Neuroscience, School of Medicine, Creighton University, Omaha, NE, United States
| | - Tithi S. Trivedi
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Jignesh Prajapati
- Department of Biochemistry & Forensic Sciences, Gujarat University, Ahmedabad, India
| | - Rakesh M. Rawal
- Department of Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Srinivas B. Muvvala
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Ray O. Bahado-Singh
- Department of Obstetrics and Gynecology, Corewell Health William Beaumont University Hospital, Royal Oak, MI, United States
| | - Senthilkumar Sadhasivam
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Yang W, Liu C, Li Z, Cui M. Exploring new drug treatment targets for immune related bone diseases using a multi omics joint analysis strategy. Sci Rep 2025; 15:10618. [PMID: 40148470 PMCID: PMC11950375 DOI: 10.1038/s41598-025-94053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
In the field of treatment and prevention of immune-related bone diseases, significant challenges persist, necessitating the urgent exploration of new and effective treatment methods. However, most existing Mendelian randomization (MR) studies are confined to a single analytical approach, which limits the comprehensive understanding of the pathogenesis and potential therapeutic targets of these diseases. In light of this, we propose the hypothesis that genetic variations in specific plasma proteins have a causal relationship with immune-related bone diseases through the MR mechanism, and that key therapeutic targets can be accurately identified using an integrated multi-omic analysis approach. This study comprehensively applied a variety of analytical methods. Firstly, the protein quantitative trait locus (pQTLs) data from two large plasma protein databases and the Genome-Wide Association Study (GWAS) data of nine immune-related bone diseases were used for Mendelian randomization (MR) analysis. At the same time, we employed the Summary-based Mendelian Randomization (SMR) method, combined with the Bayesian colocalization analysis method of coding genes, as well as the Linkage Disequilibrium Score Regression (LDSC) analysis method based on genetic correlation analysis, as methods to verify the genetic association between genes and complex diseases, thus comprehensively obtaining positive results. In addition, a Phenome-wide Association Study (PheWAS) was conducted on significantly positive genes, and their expression patterns in different tissues were also explored. Subsequently, we integrated Protein-Protein Interaction (PPI) network analysis, Gene Ontology (GO) analysis. Finally, based on the above analytical methods, drug prediction and molecular docking studies were carried out with the aim of accurately identifying key therapeutic targets. Through a comprehensive analysis using four methods, namely the Mendelian randomization (MR) analysis study, Summary-based Mendelian Randomization (SMR) analysis study, Bayesian colocalization analysis study, and Linkage Disequilibrium Score Regression (LDSC) analysis study. We found that through MR, SMR, and combined with Bayesian colocalization analysis, an association was found between rheumatoid arthritis (RA) and HDGF. Using the combination of MR and Bayesian colocalization analysis, as well as LDSC analysis, it was concluded that RA was related to CCL19 and TNFRSF14. Based on the methods of MR and Bayesian colocalization, an association was found between GPT and Crohn's disease-related arthritis, and associations were found between BTN1A1, EVI5, OGA, TNFRSF14 and multiple sclerosis (MS), and associations were found between ICAM5, CCDC50, IL17RD, UBLCP1 and psoriatic arthritis (PsA). Specifically, in the MR analysis of RA, HDGF (P_ivw = 0.0338, OR = 1.0373, 95%CI = 1.0028-1.0730), CCL19 (P_ivw = 0.0004, OR = 0.3885, 95%CI = 0.2299-0.6566), TNFRSF14 (P_ivw = 0.0007, OR = 0.6947, 95%CI = 0.5634-0.8566); in the MR analysis of MS, BTN1A1 (P_ivw = 0.0000, OR = 0.6101, 95%CI = 0.4813-0.7733), EVI5 (P_ivw = 0.0000, OR = 0.3032, 95%CI = 0.1981-0.4642), OGA (P_ivw = 0.0005, OR = 0.4599, 95%CI = 0.2966-0.7131), TNFRSF14 (P_ivw = 0.0002, OR = 0.4026, 95%CI = 0.2505-0.6471); in the MR analysis of PsA, ICAM5 (P_ivw = 0.0281, OR = 1.1742, 95%CI = 1.0174-1.3552), CCDC50 (P_ivw = 0.0092, OR = 0.7359, 95%CI = 0.5843-0.9269), IL17RD (P_ivw = 0.0006, OR = 0.7887, 95%CI = 0.6886-0.9034), UBLCP1 (P_ivw = 0.0021, OR = 0.6901, 95%CI = 0.5448-0.8741); in the MR analysis of Crohn's disease-related arthritis, GPT (P_ivw = 0.0006, OR = 0.0057, 95%CI = 0.0003-0.1111). In the Bayesian colocalization analysis of RA, HDGF (H4 = 0.8426), CCL19 (H4 = 0.9762), TNFRSF14 (H4 = 0.8016); in the Bayesian colocalization analysis of MS, BTN1A1 (H4 = 0.7660), EVI5 (H4 = 0.9800), OGA (H4 = 0.8569), TNFRSF14 (H4 = 0.8904); in the Bayesian colocalization analysis of PsA, ICAM5 (H4 = 0.9476), CCDC50 (H4 = 0.9091), IL17RD (H4 = 0.9301), UBLCP1 (H4 = 0.8862); in the Bayesian colocalization analysis of Crohn's disease-related arthritis, GPT (H4 = 0.8126). In the SMR analysis of RA, HDGF (p_SMR = 0.0338, p_HEIDI = 0.0628). In the LDSC analysis of RA, CCL19 (P = 0.0000), TNFRSF14 (P = 0.0258). By comprehensively analyzing plasma proteomic and transcriptomic data, we successfully identified key therapeutic targets for various clinical subtypes of immune-associated bone diseases. Our findings indicate that the significant positive genes associated with RA include HDGF, CCL19, and TNFRSF14; the positive gene linked to Crohn-related arthropathy is GPT; for MS, the positive genes are BTN1A1, EVI5, OGA, and TNFRSF14; and for PsA, the positive genes are ICAM5, CCDC50, IL17RD, and UBLCP1. Through this comprehensive analytical approach, we have screened potential therapeutic targets for different clinical subtypes of immune-related bone diseases. This research not only enhances our understanding of the pathogenesis of these conditions but also provides a solid theoretical foundation for subsequent drug development and clinical treatment, with the potential to yield significant advancements in the management of patients with immune-related bone diseases.
Collapse
Affiliation(s)
- Wei Yang
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chenglin Liu
- School of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zhenhua Li
- Affiliated Hospital of Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin, China.
| | - Miao Cui
- Capital Medical University, No.10, Xitoutiao, You'anmenwai, Beijing, 100069, Fengtai District, China.
| |
Collapse
|
11
|
Galande S, Ravikanth VV, Tokala RK, Satyanarayana Singh S, Rao GV, Talukdar R, Peddapulla C, Reddy DN, Sasikala M. Transcriptomic analysis of pancreatic tissue from humans and mice identifies potential gene signatures and unexplored pathways during progression from acute to chronic pancreatitis. Gene 2025; 940:149200. [PMID: 39732348 DOI: 10.1016/j.gene.2024.149200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND A comprehensive understanding of the molecular pathogenesis of chronic pancreatitis (CP), a fibroinflammatory disorder of the pancreas, is warranted for the development of targeted therapies. The current study focused on comparing the transcriptomes of pancreatic tissues obtained from patients with CP with those of two rodent models of chemically induced CP to identify dysregulated genes/signaling pathways. METHODS Pancreatitis was induced in mice using cerulein and L-arginine. Pancreatic tissues were obtained from humans and mice. The RNA was isolated, and the transcriptomes were generated using the GeneChip Human Transcriptome Array 2.0 and Clariom D Mouse Array respectively. Differentially expressed genes with log2-fold changes ≥ +2 and ≤ -2 were considered for functional and signaling pathway enrichment analysis. The expression of NUCB2, which plays a role in β-cell function, was validated by ELISA in acute pancreatitis (AP) and immune cell responses in AP and CP using flow cytometry. RESULTS The current study identifies L-arginine-induced CP as a better model for investigating the pathogenesis of human CP, with greater similarity in dysregulated genes (22%), transcription factors (34%) and enriched pathways (58%) compared to cerulein model (2%, 11% and 9%) respectively. Nesfatin-1, encoded by NUCB2, was decreased in patients with AP (12% nondiabetic, 41% post pancreatitis diabetes). The Th1 immune cell response was greater in the patients with AP (44%), whereas Th17 immune response was greater in patients with CP (18%). CONCLUSION Our study highlights potential novel and unexplored pathways involved in inflammation, fibrosis, and pain in CP and paves the way for testing them as putative drug targets using a severe disease model.
Collapse
Affiliation(s)
- Sheethal Galande
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - V V Ravikanth
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - Ranjeet K Tokala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | | | - G V Rao
- Department of Surgical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Rupjyoti Talukdar
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Chandan Peddapulla
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - D Nageshwar Reddy
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Mitnala Sasikala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India.
| |
Collapse
|
12
|
Agnoli C, Perlino F, Guerra G, Quartiroli M, Vener C, Mauri P, de Palma A, Venturelli E, Sieri S. Advanced glycation end products and breast cancer risk in a sample of the ORDET cohort. Int J Biol Markers 2025; 40:75-79. [PMID: 39834054 DOI: 10.1177/03936155241309927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
IntroductionBreast cancer is the most common cancer among women, and metabolic syndrome (MetS) is a risk factor for breast cancer, especially postmenopausal breast cancer. We evaluated the role of the advanced glycated end products (AGEs) levels contributing to the association between MetS and breast cancer risk.MethodsPlasma AGEs were measured in a case-control study nested within the Hormones and Diet in the Etiology of Breast Tumors (ORDET) cohort, including 40 incident postmenopausal breast cancer cases (20 with MetS and 20 without) and 40 postmenopausal controls (20 with MetS and 20 without). The association between AGEs and breast cancer was analyzed using Bayesian logistic regression models. An informative prior for the exposure coefficient, modeled as a normal distribution, centered on the natural logarithm of an odds ratio ((OR)=1.635) derived from prior evidence, was employed alongside weakly informative priors (WIPs). Bayesian linear regression with WIPs was used to examine the association between MetS and AGEs. Estimates were reported with SDs and 90% and 95% credible intervals (CI).ResultsAGEs were associated with higher breast cancer risk both with the informative prior (OR = 1.745, SD):0.362; 90% CI:1.218-2.390; 95% CI:1.137-2.548) and the WIP (OR = 1.861, SD = 0.661; 90% CI:1.026-3.082; 95% CI:0.924-3.528) specification. Although the difference in plasma AGEs in women with and without MetS was not significant, we found a suggestion of higher levels in women with MetS (mean difference in standardized AGEs between individuals with and without MetS = 0.155, SD = 0.245; 90% CI:-0.246 to 0.553; 95% CI:-0.322 to 0.625).ConclusionsThese data, although from a small sample of women, support a role of endogenous AGEs in the pathological pathways underlying the association between MetS and breast cancer development.
Collapse
Affiliation(s)
- Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Perlino
- Biostatistics for clinical research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Statistics, University of Warwick, Coventry, UK (current)
| | - Giulia Guerra
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Martina Quartiroli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Vener
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pierluigi Mauri
- National Research Council of Italy, Clinical Proteomics Laboratory, Elixir Infrastructure, Institute Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI), National Research Council-CNR, Naples, Italy
| | - Antonella de Palma
- National Research Council of Italy, Clinical Proteomics Laboratory, Elixir Infrastructure, Institute Biomedical Technologies, ITB-CNR, Segrate, Milan, Italy
| | - Elisabetta Venturelli
- NuMe Lab - Nutrition and Metabolomics Laboratory, Research in Nutrition and Metabolomics Unit Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
13
|
Horwitz N, Florea M, Medha KC, Liu T, Garcia V, Kim R, Lam A, Messemer K, Rios C, Almada AE, Wagers AJ. Soluble RAGE enhances muscle regeneration after cryoinjury in aged and diseased mice. PLoS One 2025; 20:e0318754. [PMID: 39999114 PMCID: PMC11856280 DOI: 10.1371/journal.pone.0318754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The Receptor for Advanced Glycation End Products (RAGE), classically considered a mediator of acute and chronic inflammatory responses, has recently been implicated by genetic knockout studies as a regulator of skeletal muscle physiology during development and following acute injury. Yet, the role of its soluble isoform, soluble RAGE (sRAGE), in muscle regeneration remains relatively unexplored. To address this knowledge gap, Adeno-Associated Virus (AAV) mediated and genetic knockin supplementation strategies were developed to specifically assess the effects of changing levels of sRAGE on muscle regeneration. We evaluated general muscle physiology and histology, including central nucleation, and myofiber size. We found that acute induction of sRAGE in aged and atherosclerotic animals accelerates muscle repair after cryoinjury. Similarly, genetic modification of the endogenous Ager gene locus to favor production of sRAGE over transmembrane RAGE accelerates repair of cryo-damaged skeletal muscle. However, increasing sRAGE via AAV delivery or using our transgenic mouse lines had no impact on muscle repair in aged or diseased mice after barium chloride (BaCl2) injury. Together, these studies identify a unique muscle regulatory activity of sRAGE that is variable across injury models and may be targeted in a context-specific manner to alter the skeletal muscle microenvironment and boost muscle regenerative output.
Collapse
Affiliation(s)
- Naftali Horwitz
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard Chemical Biology Ph.D. Program, Harvard University, Cambridge, Massachusetts, United States of America
| | - Michael Florea
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard Ph.D. Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard University, Boston, Massachusetts, United States of America
| | - K. C. Medha
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Tina Liu
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Vivian Garcia
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard Ph.D. Program in Biological and Biomedical Sciences, Division of Medical Sciences, Harvard University, Boston, Massachusetts, United States of America
| | - Rebekah Kim
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Harvard Immunology, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amy Lam
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Kathleen Messemer
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Christopher Rios
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Albert E. Almada
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Orthopaedic Surgery and Department of Stem Cell Biology and Regenerative Medicine (SCRM), University of Southern California, Los Angeles, California, United States of America
| | - Amy J. Wagers
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, Massachusetts, United States of America
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, United States of America
| |
Collapse
|
14
|
Huang Y, Zhang G, Li S, Feng J, Zhang Z. Innate and adaptive immunity in neurodegenerative disease. Cell Mol Life Sci 2025; 82:68. [PMID: 39894884 PMCID: PMC11788272 DOI: 10.1007/s00018-024-05533-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 02/04/2025]
Abstract
Neurodegenerative diseases (NDs) are a group of neurological disorders characterized by the progressive loss of selected neurons. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs. Pathologically, NDs are characterized by progressive failure of neural interactions and aberrant protein fibril aggregation and deposition, which lead to neuron loss and cognitive and behavioral impairments. Great efforts have been made to delineate the underlying mechanism of NDs. However, the very first trigger of these disorders and the state of the illness are still vague. Existing therapeutic strategies can relieve symptoms but cannot cure these diseases. The human immune system is a complex and intricate network comprising various components that work together to protect the body against pathogens and maintain overall health. They can be broadly divided into two main types: innate immunity, the first line of defense against pathogens, which acts nonspecifically, and adaptive immunity, which follows a defense process that acts more specifically and is targeted. The significance of brain immunity in maintaining the homeostatic environment of the brain, and its direct implications in NDs, has increasingly come into focus. Some components of the immune system have beneficial regulatory effects, whereas others may have detrimental effects on neurons. The intricate interplay and underlying mechanisms remain an area of active research. This review focuses on the effects of both innate and adaptive immunity on AD and PD, offering a comprehensive understanding of the initiation and regulation of brain immunity, as well as the interplay between innate and adaptive immunity in influencing the progression of NDs.
Collapse
Affiliation(s)
- Yeyu Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sheng Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jin Feng
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
15
|
Lam WLM, Gabernet G, Poth T, Sator-Schmitt M, Oquendo MB, Kast B, Lohr S, de Ponti A, Weiß L, Schneider M, Helm D, Müller-Decker K, Schirmacher P, Heikenwälder M, Klingmüller U, Schneller D, Geisler F, Nahnsen S, Angel P. RAGE is a key regulator of ductular reaction-mediated fibrosis during cholestasis. EMBO Rep 2025; 26:880-907. [PMID: 39747668 PMCID: PMC11811172 DOI: 10.1038/s44319-024-00356-7] [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: 02/28/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Ductular reaction (DR) is the hallmark of cholestatic diseases manifested in the proliferation of bile ductules lined by biliary epithelial cells (BECs). It is commonly associated with an increased risk of fibrosis and liver failure. The receptor for advanced glycation end products (RAGE) was identified as a critical mediator of DR during chronic injury. Yet, the direct link between RAGE-mediated DR and fibrosis as well as the mode of interaction between BECs and hepatic stellate cells (HSCs) to drive fibrosis remain elusive. Here, we delineate the specific function of RAGE on BECs during DR and its potential association with fibrosis in the context of cholestasis. Employing a biliary lineage tracing cholestatic liver injury mouse model, combined with whole transcriptome sequencing and in vitro analyses, we reveal a role for BEC-specific Rage activity in fostering a pro-fibrotic milieu. RAGE is predominantly expressed in BECs and contributes to DR. Notch ligand Jagged1 is secreted from activated BECs in a Rage-dependent manner and signals HSCs in trans, eventually enhancing fibrosis during cholestasis.
Collapse
Affiliation(s)
- Wai-Ling Macrina Lam
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Gisela Gabernet
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tanja Poth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Melanie Sator-Schmitt
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Morgana Barroso Oquendo
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Bettina Kast
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sabrina Lohr
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Aurora de Ponti
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Lena Weiß
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Martin Schneider
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Müller-Decker
- Tumor Models Unit, Center for Preclinical Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Doris Schneller
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Fabian Geisler
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, München, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
16
|
Jang J, Lee J, Park J, Cha S, Lee SB, Park SM, Hong SH, Kim WJ, Lee M, Yang SR. Recombinant RAGE antagonist peptide promotes alveolar epithelial cell regeneration via the RAGE/MAPKs/MMP2 pathway in emphysema. Biochem Pharmacol 2025; 231:116668. [PMID: 39608502 DOI: 10.1016/j.bcp.2024.116668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The progression of chronic obstructive pulmonary disease (COPD) results in irreversible pulmonary damage and sustained inflammatory responses. While alternative approaches have been explored, the specific role of alveolar epithelial cells in the pathogenesis of COPD remains unclear. Additionally, the association between emphysema and DAMP-RAGE signaling in COPD patients are not understood. Therefore, this study demonstrates to determine the therapeutic effect of a RAGE antagonist peptide (RAP), which we previously identified on the pathogenesis of COPD. We assessed the expression of RAGE ligands and RAGE binding signaling in COPD patients using GEO data. PPE-induced emphysema mouse model and AGER-/- mouse were employed, along treated with RAP. The association between RAGE and the development of emphysema was examined in H&E staining and western blot analysis in mouse lung tissue and BALF. We next analyzed the damage caused by oxidative stress and inflammation through CSE and RAP in human alveolar epithelial cell line A549. Our results show that inhibiting of RAGE alleviates emphysema by suppressing inflammation and MMP activity. Inhibition of RAGE in alveolar epithelial cells significantly induced the mitigation of lung injury, independent of macrophage infiltration. Furthermore, it was confirmed that RAP ameliorated CSE-induced oxidative stress, inflammation, and cell cycle arrest in human alveolar epithelial cells. These findings demonstrate that inhibiting RAGE in alveolar epithelial cells suppress lung injury and emphysema by inhibiting oxidative stress-induced inflammation and MMPs, while promoting alveolar epithelial cell proliferation. Furthermore, blocking of the DAMP-RAGE interaction through RAP offers a promising therapeutic approach for mitigating emphysema.
Collapse
Affiliation(s)
- Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jaehyun Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sangryul Cha
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea; Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon, Gangwon State, South Korea.
| |
Collapse
|
17
|
Liu X, Zheng Y, Li H, Ma Y, Cao R, Zheng Z, Tian Y, Du L, Zhang J, Zhang C, Gao J. The role of metabolites in the progression of osteoarthritis: Mechanisms and advances in therapy. J Orthop Translat 2025; 50:56-70. [PMID: 39868350 PMCID: PMC11762942 DOI: 10.1016/j.jot.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 01/28/2025] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease affected by many factors, and there is currently no effective treatment. In recent years, the latest progress in metabolomics in OA research has revealed several metabolic pathways and new specific metabolites involved in OA. Metabolites play significant roles in the identification and management of OA. This review looks back on the development history of metabolomics and the progress of this technology in OA as well as its potential clinical applications. It summarizes the applications of metabolites in the field of OA and future research directions. This understanding will advance the identification of metabolic treatment goals for OA. The translational potential of this article The development of metabolomics offers possibilities for the treatment of OA. This article reviews the relationship between metabolites associated with chondrocytes and OA. Selectively altering these three metabolic pathways and their associated metabolites may hold great potential as new focal points for OA treatment.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Yongqiang Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ruomu Cao
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuchen Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College
| | - Jinshan Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| |
Collapse
|
18
|
Krisanits BA, Kaur B, Fahey JW, Turner DP. The Anti-AGEing and RAGEing Potential of Isothiocyanates. Molecules 2024; 29:5986. [PMID: 39770075 PMCID: PMC11677037 DOI: 10.3390/molecules29245986] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/03/2025] Open
Abstract
Isothiocyanates (ITCs), found in edible plants such as cruciferous vegetables, are a group of reactive organo-sulfur phytochemicals produced by the hydrolysis of precursors known as glucosinolates. ITCs have been studied extensively both in vivo and in vitro to define their therapeutic potential for the treatment of chronic health conditions. Therapeutically, they have shown an intrinsic ability to inhibit oxidative and inflammatory phenotypes to support enhanced health. This review summarizes the current evidence supporting the observation that the antioxidant and anti-inflammatory activities of ITCs temper the pathogenic effects of a group of reactive metabolites called advanced glycation end products (AGEs). AGE exposure has significantly increased across the lifespan due to health risk factors that include dietary intake, a sedentary lifestyle, and comorbid conditions. By contributing to a chronic cycle of inflammatory stress through the aberrant activation of the transmembrane receptor for AGE (RAGE), increased AGE bioavailability is associated with chronic disease onset, progression, and severity. This review debates the potential molecular mechanisms by which ITCs may inhibit AGE bioavailability to reduce RAGE-mediated pro-oxidant and pro-inflammatory phenotypes. Bringing to light the molecular impact that ITCs may have on AGE biogenesis may stimulate novel intervention strategies for reversing or preventing the impact of lifestyle factors on chronic disease risk.
Collapse
Affiliation(s)
- Bradley A. Krisanits
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Bhoomika Kaur
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jed W. Fahey
- Departments of Medicine, Pharmacology & Molecular Sciences, Psychiatry & Behavioral Sciences, and iMIND Hopkins, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Institute of Medicine, University of Maine, Orono, ME 04469, USA
| | - David P. Turner
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA; (B.A.K.); (B.K.)
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
19
|
Radziszewski M, Galus R, Łuszczyński K, Winiarski S, Wąsowski D, Malejczyk J, Włodarski P, Ścieżyńska A. The RAGE Pathway in Skin Pathology Development: A Comprehensive Review of Its Role and Therapeutic Potential. Int J Mol Sci 2024; 25:13570. [PMID: 39769332 PMCID: PMC11676465 DOI: 10.3390/ijms252413570] [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: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, is expressed in various cell types and mediates cellular responses to a wide range of ligands. The activation of RAGE triggers complex signaling pathways that drive inflammatory, oxidative, and proliferative responses, which are increasingly implicated in the pathogenesis of skin diseases. Despite its well-established roles in conditions such as diabetes, cancer, and chronic inflammation, the contribution of RAGE to skin pathologies remains underexplored. This review synthesizes current findings on RAGE's involvement in the pathophysiology of skin diseases, including conditions such as psoriasis, atopic dermatitis, and lichen planus, focusing on its roles in inflammatory signaling, tissue remodeling, and skin cancer progression. Additionally, it examines RAGE-modulating treatments investigated in dermatological contexts, highlighting their potential as therapeutic options. Given RAGE's significance in a variety of skin conditions, further research into its mediated pathways may uncover new opportunities for targeted interventions in skin-specific RAGE signaling.
Collapse
Affiliation(s)
- Marcin Radziszewski
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
- Department of Thoracic Surgery, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Ryszard Galus
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Krzysztof Łuszczyński
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, 04-141 Warsaw, Poland
| | - Sebastian Winiarski
- Department of Thoracic Surgery, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Dariusz Wąsowski
- Department of Thoracic Surgery, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Jacek Malejczyk
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
- Institute of Health Sciences, Faculty of Medical and Health Sciences, University of Siedlce, 08-110 Siedlce, Poland
| | - Paweł Włodarski
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Aneta Ścieżyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, 04-141 Warsaw, Poland
| |
Collapse
|
20
|
Jahan H, Tufail P, Shamim S, Mohammed Khan K, Gennari M, Pizzi M, Iqbal Choudhary M. 1,2,4-Triazine derivatives as agents for the prevention of AGE-RAGE-mediated inflammatory cascade in THP-1 monocytes: An approach to prevent inflammation-induced late diabetic complications. Int Immunopharmacol 2024; 142:113145. [PMID: 39303537 DOI: 10.1016/j.intimp.2024.113145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/29/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Monocytes mainly contribute to the development and progression of vascular inflammatory conditions via the M1 polarization. The elevated levels of advanced glycation end products (AGEs) in diabetic environment lead to severe inflammation, and the release of pro-inflammatory mediators. This shifts the balance towards the pro-inflammatory state of monocytes. OBJECTIVE The current study was aimed to determine the antiglycation activity of 1,2,4-triazine derivatives, and study of their molecular basis in regulating the AGEs-mediated inflammatory responses in THP-1 monocytes. METHODS Primarily, the antiglycation activity of a series of 1,2,4-triazine derivatives was evaluated against MGO-AGEs in vitro. The toxicity of antiglycation compounds was determined by a metabolic assay, using human hepatocyte (HepG2) and monocyte (THP-1) cell lines. DCFH-DA probe was used to evaluate the antioxidant potential of the compounds. Immunocytochemistry, Western blotting, and ELISA techniques were employed to determine the levels of pro-inflammatory markers (NF-κB, RAGE, COX-1, COX-2, and PGE2) in THP-1 monocytes under in-vitro hyperglycemic conditions. RESULTS Results indicate that the triazine derivatives 22, and 23 were the most potent antiglycation agents among the entire series, while non-toxic to HepG2, and THP-1 cells. Both compounds inhibited the AGEs-induced upstream and downstream signaling of NADPH oxidase and inflammatory mediators p38 and NF-κβ, respectively, in THP-1 monocytes. They also inhibited the induction of COX-2 and its product PGE2 by suppressing AGE-RAGE interactions. Moreover, compounds 22, and 23 reversed the AGEs-mediated suppression of COX-1 in THP-1 monocytes. CONCLUSION In conclusion, 1,2,4-triazine derivatives 22, and 23 have the potential to suppress inflammatory responses under the diabetic environment through AGE-RAGE-NF-κβ/p38 nexus in THP-1 monocytes. These findings identify triazines 22, and 23 as compelling candidates for drug development, potentially beneficial for the diabetic patients with an elevated risk of vascular complications, such as atherosclerosis.
Collapse
Affiliation(s)
- Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Priya Tufail
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shahbaz Shamim
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Michele Gennari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - M Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
21
|
Simões MC, Cristóvão JS, Pardon E, Steyaert J, Fritz G, Gomes CM. Functional modulation of RAGE activation by multimeric S100B using single-domain antibodies. J Biol Chem 2024; 300:107983. [PMID: 39542249 DOI: 10.1016/j.jbc.2024.107983] [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: 07/02/2024] [Revised: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
S100B is a multifunctional protein primarily found in the brain, where it plays crucial roles in cell proliferation, differentiation, and survival. It has intracellular and extracellular functions and, depending on S100B levels, can exhibit both neurotrophic and neurotoxic activities, both mediated by the receptor for advanced glycation end products (RAGEs). Here, we report the discovery and characterization of nanobodies (Nbs) targeting dimeric and tetrameric S100B, which are the two most abundant oligomeric functional forms of the protein, aiming to modulate S100B-mediated RAGE activation. Two Nbs were selected for detailed structural and functional studies and found to bind tetrameric S100B with high affinity, as determined by biolayer interferometry (BLI) analysis and size-exclusion chromatography-stable binary complex formation. Structural and docking analyses revealed preferential contact sites of Nbs with S100B regions implicated in interactions with RAGE, namely residues at the interfacial cleft on dimeric S100B and at hydrophobic cleft formed by the association of two homodimeric units in the tetramer. In accordance, assays in SH-SY5Y cells showed that Nbs modulate the RAGE-mediated neurotrophic activity of S100B by hindering its functional interactions with the receptor. BLI competition assays between tetrameric S100B and the RAGE-VC1 domain confirmed that Nbs selectively block S100B-mediated RAGE engagement, in agreement with cell activation experiments. These findings highlight Nbs as powerful tools for elucidating molecular and cellular mechanisms through the modulation of S100B and RAGE functions, inspiring potential therapeutic applications.
Collapse
Affiliation(s)
- Margarida C Simões
- BioISI - Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Joana S Cristóvão
- BioISI - Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Günter Fritz
- Department of Cellular Microbiology, University of Hohenheim, Stuttgart, Germany
| | - Cláudio M Gomes
- BioISI - Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
22
|
Wang B, Jiang T, Qi Y, Luo S, Xia Y, Lang B, Zhang B, Zheng S. AGE-RAGE Axis and Cardiovascular Diseases: Pathophysiologic Mechanisms and Prospects for Clinical Applications. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07639-0. [PMID: 39499399 DOI: 10.1007/s10557-024-07639-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 11/07/2024]
Abstract
Advanced glycation end products (AGE), a diverse array of molecules generated through non-enzymatic glycosylation, in conjunction with the receptor of advanced glycation end products (RAGE), play a crucial role in the pathogenesis of diabetes and its associated complications. Recent studies have revealed that the AGE-RAGE axis potentially accelerated the progression of cardiovascular diseases, including heart failure, atherosclerosis, myocarditis, pulmonary hypertension, hypertension, arrhythmia, and other related conditions. The AGE-RAGE axis is intricately involved in the initiation and progression of cardiovascular diseases, independently of its engagement in diabetes. The mechanisms include oxidative stress, inflammation, alterations in autophagy flux, and mitochondrial dysfunction. Conversely, inhibition of AGE production, disruption of the binding between RAGE and its ligands, or silencing of RAGE expression could effectively impair the function of AGE-RAGE axis, thereby delaying or ameliorating the aforementioned diseases. AGE and the soluble receptor for advanced glycation end products (sRAGE) have the potential to be novel predictors of cardiovascular diseases. In this review, we provide an in-depth overview towards the biosynthetic pathway of AGE and elucidate the pathophysiological implications in various cardiovascular diseases. Furthermore, we delve into the profound role of RAGE in cardiovascular diseases, offering novel insights for further exploration of the AGE-RAGE axis and potential strategies for the prevention and management of cardiovascular disorders.
Collapse
Affiliation(s)
- Bijian Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Taidou Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Yaoyu Qi
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Sha Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Ying Xia
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Binyan Lang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Bolan Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China
| | - Shuzhan Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No.25, Taiping Street, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
23
|
Yoon T, Ahn SS, Ha JW, Ko E, Song JJ, Park YB, Lee SW. Serum Soluble Receptors for Advanced Glycation End-Products May Predict Mortality in Microscopic Polyangiitis and Granulomatosis with Polyangiitis. Yonsei Med J 2024; 65:651-660. [PMID: 39439169 PMCID: PMC11519131 DOI: 10.3349/ymj.2023.0466] [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: 11/14/2023] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 10/25/2024] Open
Abstract
PURPOSE This study aimed to investigate whether the serum extracellular newly identified receptor for advanced glycation end products binding protein (EN-RAGE) and the soluble form of RAGE (sRAGE) measured at diagnosis are associated with all-cause mortality in patients with microscopic polyangiitis (MPA) and granulomatosis with polyangiitis (GPA). MATERIALS AND METHODS Serum EN-RAGE and sRAGE were measured in 75 immunosuppressive drug-naïve MPA and GPA patients using an immunoassay, with their clinical and laboratory data reviewed. The optimal cut-off point of EN-RAGE and sRAGE was calculated by finding the threshold with the maximum sum of sensitivity and specificity. In addition, the least absolute shrinkage and selection operator regression was adopted to select variables included in the multivariable Cox proportional hazards (PH) regression model. RESULTS The median age of the patients was 67.0 years, and 34% were male. Neither serum EN-RAGE nor sRAGE at diagnosis was correlated with the Birmingham Vasculitis Activity Score. Furthermore, no correlation was observed between serum EN-RAGE and sRAGE. Deceased patients had significantly lower serum EN-RAGE and higher serum sRAGE at diagnosis compared to surviving patients. Patients with serum EN-RAGE at diagnosis ≤84.37 ng/mL and serum sRAGE at diagnosis ≥1.82 ng/mL showed significantly lower survival probabilities compared to those without. In multivariable Cox PH regression model, only serum sRAGE at diagnosis ≥1.82 ng/mL, rather than serum EN-RAGE at diagnosis ≤84.37 ng/mL, was independently associated with all-cause mortality (hazard ratio 7.094). CONCLUSION This study is the first to demonstrate that serum sRAGE at diagnosis may independently predict all-cause mortality during follow-up in patients with MPA and GPA.
Collapse
Affiliation(s)
- Taejun Yoon
- Department of Medical Science, BK21 Plus Project, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Jang Woo Ha
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eunhee Ko
- Department of Medical Science, BK21 Plus Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
24
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
25
|
Chen Q, Liu L, Ke W, Li X, Xiao H, Li Y. Association between the soluble receptor for advanced glycation end products and diabetes mellitus: systematic review and meta-analysis. BMC Endocr Disord 2024; 24:232. [PMID: 39472884 PMCID: PMC11523792 DOI: 10.1186/s12902-024-01759-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND AIMS In both type 1 diabetes (T1DM) and type 2 diabetes (T2DM), previous studies have yielded inconsistent findings regarding whether the levels of the soluble receptor for advanced glycation end products (sRAGE) are significantly altered. This meta-analysis aims to systematically evaluate the changes of sRAGE levels in patients with T1DM and T2DM. METHODS PubMed, Embase, and Web of Science were systematically searched from inception until April 2024. We included studies reporting sRAGE levels in individuals with T1DM or T2DM, using non-diabetic healthy individuals as the control group. A random-effects model was applied to conduct a meta-analysis of effect measures (means and SDs). RESULTS 49 datasets from 32 studies, involving 4948 subjects, met the inclusion criteria. A random-effects model meta-analysis showed that sRAGE levels in T1DM subjects (SMD 0.45, CI: 0.16-0.73, P = 0.002) and T2DM subjects with complications (SMD 1.59, CI: 0.77-2.41, P = 0.0001) were significantly higher than those in the control groups. No statistically significant change in sRAGE levels was observed in T2DM subjects without complications (SMD 0.01, CI: -0.61-0.64, P = 0.97). A decrease in sRAGE levels was observed in subjects with newly diagnosed T2DM (SMD-0.40, CI: -0.71- -0.09, P = 0.01). CONCLUSION This meta-analysis indicated that sRAGE levels increased in T1DM patients and T2DM patients with complications, while they decreased in newly diagnosed T2DM patients. No significant difference was observed in T2DM patients without complications. Clearly, changes in sRAGE levels in patients with T1DM or T2DM are not uniform, but depend on the different types and stages of the disease. PROSPERO REGISTRATION NUMBER CRD42024521252.
Collapse
Affiliation(s)
- Qimou Chen
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China
| | - Liehua Liu
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China
| | - Weijian Ke
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China
| | - Xuhui Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China
| | - Haipeng Xiao
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong Province, 510080, China.
| |
Collapse
|
26
|
Mougin C, Chataigner M, Lucas C, Pallet V, Bouvret E, Joffre C, Dinel AL. Dietary marine hydrolysate alleviates D-galactose-induced brain aging by attenuating cognitive alterations, oxidative stress and inflammation through the AGE-RAGE axis. PLoS One 2024; 19:e0309542. [PMID: 39446794 PMCID: PMC11500938 DOI: 10.1371/journal.pone.0309542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024] Open
Abstract
Aging represents a natural and unavoidable phenomenon in organisms. With the acceleration of population aging, investigations into aging have garnered widespread global interest. One of the most striking aspects of human aging is the decline in brain function, a phenomenon intricately tied to the onset of neurodegenerative conditions. This study aimed to assess the impact of a fish hydrolysate, rich in low-molecular-weight peptides and n-3 LC-PUFAs, on cognitive function, inflammatory response, and oxidative stress via the AGE-RAGE axis in a mouse model of accelerated aging. This model induces cognitive decline and biochemical alterations akin to those observed during natural aging. The findings revealed that fish hydrolysate exhibited a protective effect against cognitive impairment induced by D-galactose. This effect was associated with increased protein expression of SOD1 and decreased genetic expression of IL-6 and advanced glycation end products (AGE). Consequently, within the realm of preventive and personalized nutrition, fish hydrolysate emerges as a promising avenue for mitigating age-related declines in memory function.
Collapse
Affiliation(s)
- Camille Mougin
- Nutrineuro, UMR 1286, Université Bordeaux, INRAE, Bordeaux INP, Bordeaux, France
- Abyss Ingredients, Caudan, France
| | - Mathilde Chataigner
- Nutrineuro, UMR 1286, Université Bordeaux, INRAE, Bordeaux INP, Bordeaux, France
- Abyss Ingredients, Caudan, France
| | - Céline Lucas
- NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| | - Véronique Pallet
- Nutrineuro, UMR 1286, Université Bordeaux, INRAE, Bordeaux INP, Bordeaux, France
| | | | - Corinne Joffre
- Nutrineuro, UMR 1286, Université Bordeaux, INRAE, Bordeaux INP, Bordeaux, France
| | - Anne-Laure Dinel
- Nutrineuro, UMR 1286, Université Bordeaux, INRAE, Bordeaux INP, Bordeaux, France
- NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| |
Collapse
|
27
|
Xu HH, Hao SX, Sun HY, Dong XX, Lin Y, Lou H, Zhao LM, Tang PP, Dou ZJ, Han JJ, Du MH, Chen ZX, Kopylov P, Shchekochikhin D, Liu X, Zhang Y. THBru attenuates diabetic cardiomyopathy by inhibiting RAGE-dependent inflammation. Acta Pharmacol Sin 2024; 45:2107-2118. [PMID: 38862818 PMCID: PMC11420355 DOI: 10.1038/s41401-024-01307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/06/2024] [Indexed: 06/13/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a complication of diabetes mellitus characterized by heart failure and cardiac remodeling. Previous studies show that tetrahydroberberrubine (THBru) retrogrades cardiac aging by promoting PHB2-mediated mitochondrial autophagy and prevents peritoneal adhesion by suppressing inflammation. In this study we investigated whether THBru exerted protective effect against DCM in db/db mice and potential mechanisms. Eight-week-old male db/db mice were administered THBru (25, 50 mg·kg-1·d-1, i.g.) for 12 weeks. Cardiac function was assessed using echocardiography. We showed that THBru administration significantly improved both cardiac systolic and diastolic function, as well as attenuated cardiac remodeling in db/db mice. In primary neonatal mouse cardiomyocytes (NMCMs), THBru (20, 40 μM) dose-dependently ameliorated high glucose (HG)-induced cell damage, hypertrophy, inflammatory cytokines release, and reactive oxygen species (ROS) production. Using Autodock, surface plasmon resonance (SPR) and DARTS analyses, we revealed that THBru bound to the domain of the receptor for advanced glycosylation end products (RAGE), subsequently leading to inactivation of the PI3K/AKT/NF-κB pathway. Importantly, overexpression of RAGE in NMCMs reversed HG-induced inactivation of the PI3K/AKT/NF-κB pathway and subsequently counteracted the beneficial effects mediated by THBru. We conclude that THBru acts as an inhibitor of RAGE, leading to inactivation of the PI3K/AKT/NF-κB pathway. This action effectively alleviates the inflammatory responses and oxidative stress in cardiomyocytes, ultimately leading to ameliorated DCM.
Collapse
Affiliation(s)
- Heng-Hui Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Sheng-Xin Hao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - He-Yang Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Xin-Xin Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Yuan Lin
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Han Lou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Li-Min Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Ping-Ping Tang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Zi-Jia Dou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Jing-Jing Han
- Department of Pharmacy, Caoxian People's Hospital, Heze, 274400, China
| | - Meng-Han Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Zhou-Xiu Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China
| | - Philipp Kopylov
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Dmitry Shchekochikhin
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China.
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China.
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150000, China.
- State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, 150000, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150000, China.
| |
Collapse
|
28
|
Diao Z, Guo D, Zhang J, Zhang R, Li C, Chen H, Ma Y. Causal relationship between modifiable risk factors and knee osteoarthritis: a Mendelian randomization study. Front Med (Lausanne) 2024; 11:1405188. [PMID: 39286647 PMCID: PMC11402680 DOI: 10.3389/fmed.2024.1405188] [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: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 09/19/2024] Open
Abstract
Background While several risk factors for knee osteoarthritis (KOA) have been recognized, the pathogenesis of KOA and the causal relationship between modifiable risk factors and KOA in genetic epidemiology remain unclear. This study aimed to determine the causal relationship between KOA and its risk factors. Methods Data were obtained from published Genome-Wide Association study (GWAS) databases. A two-sample Mendelian randomization (MR) analysis was performed with genetic variants associated with risk factors as instrumental variables and KOA as outcome. First, inverse variance weighting was used as the main MR analysis method, and then a series of sensitivity analyses were conducted to comprehensively evaluate the causal relationship between them. Results Univariate forward MR analysis revealed that genetically predicted hypothyroidism, hyperthyroidism/thyrotoxicosis, educational level, income level, metabolic syndrome (MS), essential hypertension, height, hot drink temperature, diet (abstaining from sugar-sweetened or wheat products), and psychological and psychiatric disorders (stress, depression, and anxiety) were causally associated with KOA. Reverse MR exhibits a causal association between KOA and educational attainment. Multivariate MR analysis adjusted for the inclusion of potential mediators, such as body mass index (BMI), smoking, alcohol consumption, and sex, exhibited some variation in causal effects. However, hyperthyroidism/thyrotoxicosis had a significant causal effect on KOA, and there was good evidence that height, hypothyroidism, educational level, psychological and psychiatric disorders (stress, depression, and anxiety), and abstaining from wheat products had an independent causal relationship. The mediating effect of BMI as a mediator was also identified. Conclusion This study used MR to validate the causal relationship between KOA and its risk factors, providing new insights for preventing and treating KOA in clinical practice and for developing public health policies.
Collapse
Affiliation(s)
- Zhihao Diao
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Danyang Guo
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingzhi Zhang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruiyu Zhang
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunjing Li
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Chen
- Complutense University of Madrid, Madrid, Spain
| | - Yuxia Ma
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Connolly DM, Madden LA, Edwards VC, Lee VM. Brain and Lung Biomarker Responses to Hyperoxic Hypobaric Decompression. Aerosp Med Hum Perform 2024; 95:667-674. [PMID: 39169490 DOI: 10.3357/amhp.6391.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
INTRODUCTION: Biomarker responses to intensive decompression indicate systemic proinflammatory responses and possible neurological stress. To further investigate responses, 12 additional brain and lung biomarkers were assayed.METHODS: A total of 15 healthy men (20 to 50 yr) undertook consecutive same-day ascents to 25,000 ft (7620 m), following denitrogenation, breathing 100% oxygen. Venous blood was sampled at baseline (T0), after the second ascent (T8), and next morning (T24). Soluble protein markers of brain and lung insult were analyzed by enzyme-linked immunosorbent assay with plasma microparticles quantified using flow cytometry.RESULTS: Levels of monocyte chemoattractant protein-1 and high mobility group box protein 1 were elevated at T8, by 36% and 16%, respectively, before returning to baseline. Levels of soluble receptor for advanced glycation end products fell by 8%, recovering by T24. Brain-derived neurotrophic factor rose by 80% over baseline at T24. Monocyte microparticle levels rose by factors of 3.7 at T8 and 2.7 at T24 due to early and late responses in different subjects. Other biomarkers were unaffected or not detected consistently.DISCUSSION: The elevated biomarkers at T8 suggest a neuroinflammatory response, with later elevation of brain-derived neurotrophic factor at T24 indicating an ongoing neurotrophic response and incomplete recovery. A substantial increase at T8 in the ratio of high mobility group box protein 1 to soluble receptor for advanced glycation end products suggests this axis may mediate the systemic inflammatory response to decompression. The mechanism of neuroinflammation is unclear but elevation of monocyte microparticles and monocyte chemoattractant protein-1 imply a key role for activated monocytes and/or macrophages.Connolly DM, Madden LA, Edwards VC, Lee VM. Brain and lung biomarker responses to hyperoxic hypobaric decompression. Aerosp Med Hum Perform. 2024; 95(9):667-674.
Collapse
|
30
|
Galli F, Bartolini D, Ronco C. Oxidative stress, defective proteostasis and immunometabolic complications in critically ill patients. Eur J Clin Invest 2024; 54:e14229. [PMID: 38676423 DOI: 10.1111/eci.14229] [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: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Oxidative stress (OS) develops in critically ill patients as a metabolic consequence of the immunoinflammatory and degenerative processes of the tissues. These induce increased and/or dysregulated fluxes of reactive species enhancing their pro-oxidant activity and toxicity. At the same time, OS sustains its own inflammatory and immunometabolic pathogenesis, leading to a pervasive and vitious cycle of events that contribute to defective immunity, organ dysfunction and poor prognosis. Protein damage is a key player of these OS effects; it generates increased levels of protein oxidation products and misfolded proteins in both the cellular and extracellular environment, and contributes to forms DAMPs and other proteinaceous material to be removed by endocytosis and proteostasis processes of different cell types, as endothelial cells, tissue resident monocytes-macrophages and peripheral immune cells. An excess of OS and protein damage in critical illness can overwhelm such cellular processes ultimately interfering with systemic proteostasis, and consequently with innate immunity and cell death pathways of the tissues thus sustaining organ dysfunction mechanisms. Extracorporeal therapies based on biocompatible/bioactive membranes and new adsorption techniques may hold some potential in reducing the impact of OS on the defective proteostasis of patients with critical illness. These can help neutralizing reactive and toxic species, also removing solutes in a wide spectrum of molecular weights thus improving proteostasis and its immunometabolic corelates. Pharmacological therapy is also moving steps forward which could help to enhance the efficacy of extracorporeal treatments. This narrative review article explores the aspects behind the origin and pathogenic role of OS in intensive care and critically ill patients, with a focus on protein damage as a cause of impaired systemic proteostasis and immune dysfunction in critical illness.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Ronco
- Department of Medicine, International Renal Research Institute of Vicenza, University of Padova, San Bortolo Hospital Vicenza, Vicenza, Italy
| |
Collapse
|
31
|
Li K, Leng Y, Lei D, Zhang H, Ding M, Lo WLA. Causal link between metabolic related factors and osteoarthritis: a Mendelian randomization investigation. Front Nutr 2024; 11:1424286. [PMID: 39206315 PMCID: PMC11349640 DOI: 10.3389/fnut.2024.1424286] [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: 04/27/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Metabolic syndrome (MetS) is significantly associated with osteoarthritis (OA), especially in MetS patients with blood glucose abnormalities, such as elevated fasting blood glucose (FG), which may increase OA risk. Dietary modifications, especially the intake of polyunsaturated fatty acids (PUFAs), are regarded as a potential means of preventing MetS and its complications. However, regarding the effects of FG, Omega-3s, and Omega-6s on OA, the research conclusions are conflicting, which is attributed to the complexity of the pathogenesis of OA. Therefore, it is imperative to thoroughly evaluate multiple factors to fully understand their role in OA, which needs further exploration and clarification. Methods Two-sample univariable Mendelian randomization (UVMR) and multivariable Mendelian randomization (MVMR) were employed to examine the causal effect of metabolic related factors on hip OA (HOA) or knee OA (KOA). The exposure and outcome datasets were obtained from Open GWAS IEU. All cases were independent European ancestry data. Three MR methods were performed to estimate the causal effect: inverse-variance weighting (IVW), weighted median method (WMM), and MR-Egger regression. Additionally, the intercept analysis in MR-Egger regression is used to estimate pleiotropy, and the IVW method and MR-Egger regression are used to test the heterogeneity. Results The UVMR analysis revealed a causal relationship between FG and HOA. By MVMR analysis, the study discovered a significant link between FG (OR = 0.79, 95%CI: 0.64∼0.99, p = 0.036) and KOA after accounting for body mass index (BMI), age, and sex hormone-binding globulin (SHBG). However, no causal effects of FG on HOA were seen. Omega-3s and Omega-6s did not have a causal influence on HOA or KOA. No significant evidence of pleiotropy was identified. Discussion The MR investigation showed a protective effect of FG on KOA development but no causal relationship between FG and HOA. No causal effect of Omega-3s and Omega-6s on HOA and KOA was observed. Shared genetic overlaps might also exist between BMI and age, SHBG and PUFAs for OA development. This finding offers a novel insight into the treatment and prevention of KOA from glucose metabolism perspective. The FG cutoff value should be explored in the future, and consideration should be given to demonstrating the study in populations other than Europeans.
Collapse
Affiliation(s)
- Kai Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Leng
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Lei
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haojie Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minghui Ding
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wai Leung Ambrose Lo
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering and Technology Research Centre for Rehabilitation Medicine and Translation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
32
|
Zhou M, Zhang Y, Shi L, Li L, Zhang D, Gong Z, Wu Q. Activation and modulation of the AGEs-RAGE axis: Implications for inflammatory pathologies and therapeutic interventions - A review. Pharmacol Res 2024; 206:107282. [PMID: 38914383 DOI: 10.1016/j.phrs.2024.107282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/26/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Chronic inflammation is a common foundation for the development of many non-communicable diseases, particularly diabetes, atherosclerosis, and tumors. The activation of the axis involving Advanced Glycation End products (AGEs) and their receptor RAGE is a key promotive factor in the chronic inflammation process, influencing the pathological progression of these diseases. The accumulation of AGEs in the body results from an increase in glycation reactions and oxidative stress, especially pronounced in individuals with diabetes. By binding to RAGE, AGEs activate signaling pathways such as NF-κB, promoting the release of inflammatory factors, exacerbating cell damage and inflammation, and further advancing the formation of atherosclerotic plaques and tumor development. This review will delve into the molecular mechanisms by which the AGEs-RAGE axis activates chronic inflammation in the aforementioned diseases, as well as strategies to inhibit the AGEs-RAGE axis, aiming to slow or halt the progression of chronic inflammation and related diseases. This includes the development of AGEs inhibitors, RAGE antagonists, and interventions targeting upstream and downstream signaling pathways. Additionally, the early detection of AGEs levels and RAGE expression as biomarkers provides new avenues for the prevention and treatment of diabetes, atherosclerosis, and tumors.
Collapse
Affiliation(s)
- Mengzhou Zhou
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Yuyan Zhang
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Lin Shi
- Wuhan Caidian District Public Inspection and Testing Center, Wuhan, Hubei 430068, PR China
| | - Liangchao Li
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Duo Zhang
- Hubei Standardization and Quality Institute, Wuhan,Hubei 430068, PR China
| | - Zihao Gong
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China
| | - Qian Wu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| |
Collapse
|
33
|
Peixoto DO, Bittencourt RR, Gasparotto J, Kessler FGC, Brum PO, Somensi N, Girardi CS, Dos Santos da Silva L, Outeiro TF, Moreira JCF, Gelain DP. Increased alpha-synuclein and neuroinflammation in the substantia nigra triggered by systemic inflammation are reversed by targeted inhibition of the receptor for advanced glycation end products (RAGE). J Neurochem 2024; 168:1587-1607. [PMID: 37661637 DOI: 10.1111/jnc.15956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a protein of the immunoglobulin superfamily capable of regulating inflammation. Considering the role of this receptor in the initiation and establishment of neuroinflammation, and the limited understanding of the function of RAGE in the maintenance of this condition, this study describes the effects of RAGE inhibition in the brain, through an intranasal treatment with the antagonist FPS-ZM1, in an animal model of chronic neuroinflammation induced by acute intraperitoneal injection of lipopolysaccharide (LPS). Seventy days after LPS administration (2 mg/kg, i.p.), Wistar rats received, intranasally, 1.2 mg of FPS-ZM1 over 14 days. On days 88 and 89, the animals were submitted to the open-field test and were killed on day 90 after the intraperitoneal injection of LPS. Our results indicate that blockade of encephalic RAGE attenuates LPS-induced chronic neuroinflammation in different brain regions. Furthermore, we found that intranasal FPS-ZM1 administration reduced levels of gliosis markers, RAGE ligands, and α-synuclein in the substantia nigra pars compacta. Additionally, the treatment also reversed the increase in S100 calcium-binding protein B (RAGE ligand) in the cerebrospinal fluid and the cognitive-behavioral deficits promoted by LPS-less time spent in the central zone of the open-field arena (more time in the lateral zones), decreased total distance traveled, and increased number of freezing episodes. In summary, our study demonstrates the prominent role of RAGE in the maintenance of a chronic neuroinflammatory state triggered by a single episode of systemic inflammation and also points to possible future RAGE-based therapeutic approaches to treat conditions in which chronic neuroinflammation and increased α-synuclein levels could play a relevant role, such as in Parkinson's disease.
Collapse
Affiliation(s)
- Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Alicante, Spain
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (ICB-UNIFAL), Alfenas, Brazil
| | - Flávio Gabriel Carazza Kessler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | | | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Carolina Saibro Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Lucas Dos Santos da Silva
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Natural Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| |
Collapse
|
34
|
Gasparotto J, Somensi N, Girardi CS, Bittencourt RR, de Oliveira LM, Hoefel LP, Scheibel IM, Peixoto DO, Moreira JCF, Outeiro TF, Gelain DP. Is it all the RAGE? Defining the role of the receptor for advanced glycation end products in Parkinson's disease. J Neurochem 2024; 168:1608-1624. [PMID: 37381043 DOI: 10.1111/jnc.15890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a transmembrane receptor that belongs to the immunoglobulin superfamily and is extensively associated with chronic inflammation in non-transmissible diseases. As chronic inflammation is consistently present in neurodegenerative diseases, it was largely assumed that RAGE could act as a critical modulator of neuroinflammation in Parkinson's disease (PD), similar to what was reported for Alzheimer's disease (AD), where RAGE is postulated to mediate pro-inflammatory signaling in microglia by binding to amyloid-β peptide. However, accumulating evidence from studies of RAGE in PD models suggests a less obvious scenario. Here, we review physiological aspects of RAGE and address the current questions about the potential involvement of this receptor in the cellular events that may be critical for the development and progression of PD, exploring possible mechanisms beyond the classical view of the microglial activation/neuroinflammation/neurodegeneration axis that is widely assumed to be the general mechanism of RAGE action in the adult brain.
Collapse
Affiliation(s)
- Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brazil
| | - Nauana Somensi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carolina Saibro Girardi
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Reykla Ramon Bittencourt
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Martinewski de Oliveira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laura Piloneto Hoefel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ingrid Matsubara Scheibel
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Oppermann Peixoto
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Claudio Fonseca Moreira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Daniel Pens Gelain
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
35
|
Niri P, Saha A, Polopalli S, Kumar M, Das S, Chattopadhyay P. Role of biomarkers and molecular signaling pathways in acute lung injury. Fundam Clin Pharmacol 2024; 38:640-657. [PMID: 38279523 DOI: 10.1111/fcp.12987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/07/2023] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is caused by bacterial, fungal, and viral infections. When pathogens invade the lungs, the immune system responds by producing cytokines, chemokines, and interferons to promote the infiltration of phagocytic cells, which are essential for pathogen clearance. Their excess production causes an overactive immune response and a pathological hyper-inflammatory state, which leads to ALI. Until now, there is no particular pharmaceutical treatment available for ALI despite known inflammatory mediators like neutrophil extracellular traps (NETs) and reactive oxygen species (ROS). OBJECTIVES Therefore, the primary objective of this review is to provide the clear overview on the mechanisms controlling NETs, ROS formation, and other relevant processes during the pathogenesis of ALI. In addition, we have discussed the significance of epithelial and endothelial damage indicators and several molecular signaling pathways associated with ALI. METHODS The literature review was done from Web of Science, Scopus, PubMed, and Google Scholar for ALI, NETs, ROS, inflammation, biomarkers, Toll- and nucleotide-binding oligomerization domain (NOD)-like receptors, alveolar damage, pro-inflammatory cytokines, and epithelial/endothelial damage alone or in combination. RESULTS This review summarized the main clinical signs of ALI, including the regulation and distinct function of epithelial and endothelial biomarkers, NETs, ROS, and pattern recognition receptors (PRRs). CONCLUSION However, no particular drugs including vaccine for ALI has been established. Furthermore, there is a lack of validated diagnostic tools and a poor predictive rationality of current therapeutic biomarkers. Hence, extensive and precise research is required to speed up the process of drug testing and development by the application of artificial intelligence technologies, structure-based drug design, in-silico approaches, and drug repurposing.
Collapse
Affiliation(s)
- Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Subramanyam Polopalli
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, India
| | - Sanghita Das
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
- Department of Chemical Technology, University of Calcutta, Kolkata, 700009, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, 784 001, India
| |
Collapse
|
36
|
Kanikowska D, Kanikowska A, Strojny Z, Kawka E, Zawada A, Rutkowski R, Litwinowicz M, Sato M, Grzymisławski M, Bręborowicz A, Witowski J, Korybalska K. Assessment of EN-RAGE, sRAGE, and its isoforms: cRAGE, esRAGE in obese patients treated by moderate caloric restriction combined with physical activity conducted in hospital condition. Cytokine 2024; 180:156665. [PMID: 38823153 DOI: 10.1016/j.cyto.2024.156665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND AGEs, their receptor (RAGE), and the extracellular newly identified receptor for AGEs product-binding protein (EN-RAGE) are implicated in the pathogenesis of inflammation. AIM We analyzed serum EN-RAGE, soluble RAGE (sRAGE), and their isoforms: endogenous secretory - esRAGE and cleaved - cRAGE concentrations in lean controls (n = 74) and in patients with obesity (n = 71) treated for three weeks with moderate calorie restriction (CR) combined with physical activity in a hospital condition. METHODS Using the ELISA method, serum sRAGE, esRAGE, and EN-RAGE were measured before and after CR. RESULTS The serum level of sRAGE and esRAGE in patients with obesity was lower than that in non-obese individuals, contrary to cRAGE. EN-RAGE concentration was about three times higher in obese patients. Gradually, a rise in BMI resulted in sRAGE, esRAGE reduction, and EN-RAGE increase. The sRAGE concentration was sex-dependent, indicating a higher value in lean men. A moderate negative correlation was observed between BMI and all RAGE isoforms, whereas EN-RAGE displays a positive correlation. CR resulted in an expected decrease in anthropometric, metabolic, and proinflammatory parameters and EN-RAGE, but no RAGE isoforms. The ratio EN-RAGE/sRAGE was higher in obese humans than in control and was not modified by CR. CONCLUSION Obesity decreases sRAGE and esRAGE and increases EN-RAGE concentration. Moderate CR and physical activity by decreasing inflammation reduces EN-RAGE but is insufficient to increase sRAGE and esRAGE to the extent observed in lean patients. EN-RAGE instead of sRAGE could be helpful to indicate a better outcome of moderate dietary intervention in obese subjects.
Collapse
Affiliation(s)
- Dominika Kanikowska
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland.
| | - Alina Kanikowska
- Department of Internal Diseases, Metabolism and Nutrition, Poznań University of Medical Science, Poznań, Poland
| | - Zofia Strojny
- Department of Conservative Dentistry and Endodontics, Poznań University of Medical Sciences, Poznań, Poland
| | - Edyta Kawka
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Agnieszka Zawada
- Department of Internal Diseases, Metabolism and Nutrition, Poznań University of Medical Science, Poznań, Poland
| | - Rafał Rutkowski
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Monika Litwinowicz
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Maki Sato
- Institutional Research, Aichi Medical University School of Medicine, Aichi, Japan
| | - Marian Grzymisławski
- Department of Internal Diseases, Metabolism and Nutrition, Poznań University of Medical Science, Poznań, Poland
| | - Andrzej Bręborowicz
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland; Collegium Medicum, Zielona Góra University, Zielona Góra, Poland
| | - Janusz Witowski
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Korybalska
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
37
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
38
|
Lian W, Wang Z, Zhou F, Yuan X, Xia C, Wang W, Yan Y, Cheng Y, Yang H, Xu J, He J, Zhang W. Cornuside ameliorates cognitive impairments via RAGE/TXNIP/NF-κB signaling in Aβ 1-42 induced Alzheimer's disease mice. J Neuroimmune Pharmacol 2024; 19:24. [PMID: 38780885 DOI: 10.1007/s11481-024-10120-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/21/2024] [Indexed: 05/25/2024]
Abstract
Cornuside has been discovered to improve learning and memory in AD mice, however, its underlying mechanism was not fully understood. In the present study, we established an AD mice model by intracerebroventricular injection of Aβ1-42, which were treated with cornuside (3, 10, 30 mg/kg) for 2 weeks. Cornuside significantly ameliorated cognitive function of AD mice in series of behavioral tests, including Morris water maze test, nest building test, novel object recognition test and step-down test. Additionally, cornuside could attenuate neuronal injury, and promote cholinergic synaptic transmission by restoring the level of acetylcholine (ACh) via inhibiting acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), as well as facilitating choline acetyltransferase (ChAT). Furthermore, cornuside inhibited oxidative stress levels amplified as decreased malondialdehyde (MDA), by inhibiting TXNIP expression, improving total anti-oxidative capacity (TAOC), raising activities of superoxide dismutase (SOD) and catalase (CAT). Cornuside also reduced the activation of microglia and astrocytes, decreased the level of proinflammatory factors TNF-α, IL-6, IL-1β, iNOS and COX2 via interfering RAGE-mediated IKK-IκB-NF-κB phosphorylation. Similar anti-oxidative and anti-inflammatory effects were also found in LPS-stimulated BV2 cells via hampering RAGE-mediated TXNIP activation and NF-κB nuclear translocation. Virtual docking revealed that cornuside could interact with the active pocket of RAGE V domain directly. In conclusion, cornuside could bind to the RAGE directly impeding the interaction of Aβ and RAGE, and cut down the expression of TXNIP inhibiting ROS production and oxidative stress, as well as hamper NF-κB p65 mediated the inflammation.
Collapse
Grants
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- ZRJY2023-QM10, ZRJY2021-BJ06, ZRJY2021-TD06, ZRJY2021-QM16, ZRJY2023-QM28 Elite Medical Professionals Project of China-Japan Friendship Hospital
- 3332023096 Central Universities Fundamental for Basic Scientific Research of Peking Union Medical College
- 2022SLZDCY-001 Yan'an Science and Technology Plan Project
- 2022JZ-49 Key Project Funding for Shaanxi Provincial Natural Science Basic Rearch Program
- 82273809, 82273815, 82073731 National Natural Science Foundation of China
- 82273809, 82273815, 82073731 National Natural Science Foundation of China
- 2023-NHLHCRF-CXYW-01, 2022-NHLHCRF-YNZY-01 National High Level hospital Clinical Research Funding
- 2023-NHLHCRF-CXYW-01, 2022-NHLHCRF-YNZY-01 National High Level hospital Clinical Research Funding
- 2022-JKCS-16 Nonprofit Central Research Institute Fund of Chinese Academy of Medical Science
- CPA-B04-ZC-2021-005 Chinese Pharmaceutical Association-Yiling Biomedical Innovation Fund Project
Collapse
Affiliation(s)
- Wenwen Lian
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Zexing Wang
- School of Life Science, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Fulin Zhou
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Xiaotang Yuan
- School of Life Science, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Congyuan Xia
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Wenping Wang
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Yu Yan
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Yunchi Cheng
- Department of Pharmacology, School of Medicine, Yale University, Connecticut, New Haven, USA
| | - Hua Yang
- School of Chemistry and Chemical Engineering, Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy & New Function Materials, Yan'an University, Yan'an, Shaanxi, 716000, People's Republic of China
| | - Jiekun Xu
- School of Life Science, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.
| | - Jun He
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China.
| | - Weiku Zhang
- Department of Pharmacy & Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China.
| |
Collapse
|
39
|
Grauen Larsen H, Sun J, Sjögren M, Borné Y, Engström G, Nilsson P, Orho-Melander M, Goncalves I, Nilsson J, Melander O, Schiopu A. The Gly82Ser polymorphism in the receptor for advanced glycation endproducts increases the risk for coronary events in the general population. Sci Rep 2024; 14:11567. [PMID: 38773223 PMCID: PMC11109115 DOI: 10.1038/s41598-024-62385-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/16/2024] [Indexed: 05/23/2024] Open
Abstract
The receptor for advanced glycation endproducts (RAGE) has pro-inflammatory and pro-atherogenic effects. Low plasma levels of soluble RAGE (sRAGE), a decoy receptor for RAGE ligands, have been associated with increased risk for major adverse coronary events (MACE) in the general population. We performed a genome-wide association study to identify genetic determinants of plasma sRAGE in 4338 individuals from the cardiovascular arm of the Malmö Diet and Cancer study (MDC-CV). Further, we explored the associations between these genetic variants, incident first-time MACE and mortality in 24,640 unrelated individuals of European ancestry from the MDC cohort. The minor alleles of four single nucleotide polymorphisms (SNPs): rs2070600, rs204993, rs116653040, and rs7306778 were independently associated with lower plasma sRAGE. The minor T (vs. C) allele of rs2070600 was associated with increased risk for MACE [HR 1.13 95% CI (1.02-1.25), P = 0.016]. Neither SNP was associated with mortality. This is the largest study to demonstrate a link between a genetic sRAGE determinant and CV risk. Only rs2070600, which enhances RAGE function by inducing a Gly82Ser polymorphism in the ligand-binding domain, was associated with MACE. The lack of associations with incident MACE for the other sRAGE-lowering SNPs suggests that this functional RAGE modification is central for the observed relationship.
Collapse
Affiliation(s)
- Helena Grauen Larsen
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital Malmö, 21428, Malmö, Sweden
| | - Jiangming Sun
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Marketa Sjögren
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Yan Borné
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Peter Nilsson
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital Malmö, 21428, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, 21428, Malmö, Sweden
- Department of Internal Medicine, Skane University Hospital Lund, 22242, Lund, Sweden
| | - Alexandru Schiopu
- Department of Internal Medicine, Skane University Hospital Lund, 22242, Lund, Sweden.
- Department of Translational Medicine, Lund University, 21428, Malmö, Sweden.
- Nicolae Simionescu Institute of Cellular Biology and Pathology, 050568, Bucharest, Romania.
| |
Collapse
|
40
|
Chang NP, DaPrano EM, Lindman M, Estevez I, Chou TW, Evans WR, Nissenbaum M, McCourt M, Alzate D, Atkins C, Kusnecov AW, Huda R, Daniels BP. Neuronal DAMPs exacerbate neurodegeneration via astrocytic RIPK3 signaling. JCI Insight 2024; 9:e177002. [PMID: 38713518 PMCID: PMC11382884 DOI: 10.1172/jci.insight.177002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
Astrocyte activation is a common feature of neurodegenerative diseases. However, the ways in which dying neurons influence the activity of astrocytes is poorly understood. Receptor interacting protein kinase-3 (RIPK3) signaling has recently been described as a key regulator of neuroinflammation, but whether this kinase mediates astrocytic responsiveness to neuronal death has not yet been studied. Here, we used the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine model of Parkinson's disease to show that activation of astrocytic RIPK3 drives dopaminergic cell death and axon damage. Transcriptomic profiling revealed that astrocytic RIPK3 promoted gene expression associated with neuroinflammation and movement disorders, and this coincided with significant engagement of damage-associated molecular pattern signaling. In mechanistic experiments, we showed that factors released from dying neurons signaled through receptor for advanced glycation endproducts to induce astrocytic RIPK3 signaling, which conferred inflammatory and neurotoxic functional activity. These findings highlight a mechanism of neuron-glia crosstalk in which neuronal death perpetuates further neurodegeneration by engaging inflammatory astrocyte activation via RIPK3.
Collapse
Affiliation(s)
| | | | | | | | | | - Wesley R Evans
- Department of Cell Biology and Neuroscience
- W. M. Keck Center for Collaborative Neuroscience, and
| | | | | | | | | | | | - Rafiq Huda
- Department of Cell Biology and Neuroscience
- W. M. Keck Center for Collaborative Neuroscience, and
| | | |
Collapse
|
41
|
Chen RJ, Nabila A, Gal Toth J, Stuhlmann H, Toth M. The chemokine XCL1 functions as a pregnancy hormone to program offspring innate anxiety. Brain Behav Immun 2024; 118:178-189. [PMID: 38428650 PMCID: PMC11044916 DOI: 10.1016/j.bbi.2024.02.032] [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: 07/28/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Elevated levels of cytokines in maternal circulation increase the offspring's risk for neuropsychiatric disease. Because of their low homeostatic levels, circulating maternal cytokines during normal pregnancies have not been considered to play a role in fetal brain development and offspring behavior. Here we report that the T/NK cell chemotactic cytokine XCL1, a local paracrine immune signal, can function as a pregnancy hormone and is required for the proper development of placenta and male offspring approach-avoidance behavior. We found that circulating XCL1 levels were at a low pregestational level throughout pregnancy except for a midgestational rise and fall. Blunted elevation in maternal plasma XCL1 in dams with a genetic 5HT1A receptor deficit or following neutralization by anti-XCL1 antibodies increased the expression of tissue damage associated factors in WT fetal placenta and led to increased innate anxiety and stress reactivity in the WT male offspring. Therefore, chemokines like XCL1 may act as pregnancy hormones to regulate placenta development and offspring emotional behavior.
Collapse
Affiliation(s)
- Rosa J Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anika Nabila
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Judit Gal Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi Stuhlmann
- Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miklos Toth
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
42
|
Seow SR, Mat S, Ahmad Azam A, Rajab NF, Safinar Ismail I, Singh DKA, Shahar S, Tan MP, Berenbaum F. Impact of diabetes mellitus on osteoarthritis: a scoping review on biomarkers. Expert Rev Mol Med 2024; 26:e8. [PMID: 38606593 PMCID: PMC11062141 DOI: 10.1017/erm.2024.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 01/22/2024] [Accepted: 02/22/2024] [Indexed: 04/13/2024]
Abstract
Osteoarthritis (OA) commonly affects the knee and hip joints and accounts for 19.3% of disability-adjusted life years and years lived with disability worldwide (Refs , ). Early management is important in order to avoid disability uphold quality of life (Ref. ). However, a lack of awareness of subclinical and early symptomatic stages of OA often hampers early management (Ref. ). Moreover, late diagnosis of OA among those with severe disease, at a stage when OA management becomes more complicated is common (Refs , , , ). Established risk factors for the development and progression of OA include increasing age, female, history of trauma and obesity (Ref. ). Recent studies have also drawn a link between OA and metabolic syndrome, which is characterized by insulin resistance, dyslipidaemia and hypertension (Refs , ).
Collapse
Affiliation(s)
- Shi Rui Seow
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sumaiyah Mat
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amalina Ahmad Azam
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Intan Safinar Ismail
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Devinder Kaur Ajit Singh
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Suzana Shahar
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Maw Pin Tan
- ACT4Health Services and Consultancy Sdn. Bhd, Petaling Jaya, Selangor, Malaysia
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Francis Berenbaum
- Rheumatology, Saint-Antoine Hospital, AP-HP, INSERM CSRA, Sorbonne Université, Paris, France
| |
Collapse
|
43
|
Castro-Gomez S, Heneka MT. Innate immune activation in neurodegenerative diseases. Immunity 2024; 57:790-814. [PMID: 38599171 DOI: 10.1016/j.immuni.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Activation of the innate immune system following pattern recognition receptor binding has emerged as one of the major pathogenic mechanisms in neurodegenerative disease. Experimental, epidemiological, pathological, and genetic evidence underscores the meaning of innate immune activation during the prodromal as well as clinical phases of several neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia. Importantly, innate immune activation and the subsequent release of inflammatory mediators contribute mechanistically to other hallmarks of neurodegenerative diseases such as aberrant proteostatis, pathological protein aggregation, cytoskeleton abnormalities, altered energy homeostasis, RNA and DNA defects, and synaptic and network disbalance and ultimately to the induction of neuronal cell death. In this review, we discuss common mechanisms of innate immune activation in neurodegeneration, with particular emphasis on the pattern recognition receptors (PRRs) and other receptors involved in the detection of damage-associated molecular patterns (DAMPs).
Collapse
Affiliation(s)
- Sergio Castro-Gomez
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany; Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
44
|
Yehya N, Booth TJ, Ardhanari GD, Thompson JM, Lam LM, Till JE, Mai MV, Keim G, McKeone DJ, Halstead ES, Lahni P, Varisco BM, Zhou W, Carpenter EL, Christie JD, Mangalmurti NS. Inflammatory and tissue injury marker dynamics in pediatric acute respiratory distress syndrome. J Clin Invest 2024; 134:e177896. [PMID: 38573766 PMCID: PMC11093602 DOI: 10.1172/jci177896] [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: 11/22/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUNDThe molecular signature of pediatric acute respiratory distress syndrome (ARDS) is poorly described, and the degree to which hyperinflammation or specific tissue injury contributes to outcomes is unknown. Therefore, we profiled inflammation and tissue injury dynamics over the first 7 days of ARDS, and associated specific biomarkers with mortality, persistent ARDS, and persistent multiple organ dysfunction syndrome (MODS).METHODSIn a single-center prospective cohort of intubated pediatric patients with ARDS, we collected plasma on days 0, 3, and 7. Nineteen biomarkers reflecting inflammation, tissue injury, and damage-associated molecular patterns (DAMPs) were measured. We assessed the relationship between biomarkers and trajectories with mortality, persistent ARDS, or persistent MODS using multivariable mixed effect models.RESULTSIn 279 patients (64 [23%] nonsurvivors), hyperinflammatory cytokines, tissue injury markers, and DAMPs were higher in nonsurvivors. Survivors and nonsurvivors showed different biomarker trajectories. IL-1α, soluble tumor necrosis factor receptor 1, angiopoietin 2 (ANG2), and surfactant protein D increased in nonsurvivors, while DAMPs remained persistently elevated. ANG2 and procollagen type III N-terminal peptide were associated with persistent ARDS, whereas multiple cytokines, tissue injury markers, and DAMPs were associated with persistent MODS. Corticosteroid use did not impact the association of biomarker levels or trajectory with mortality.CONCLUSIONSPediatric ARDS survivors and nonsurvivors had distinct biomarker trajectories, with cytokines, endothelial and alveolar epithelial injury, and DAMPs elevated in nonsurvivors. Mortality markers overlapped with markers associated with persistent MODS, rather than persistent ARDS.FUNDINGNIH (K23HL-136688, R01-HL148054).
Collapse
Affiliation(s)
- Nadir Yehya
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia and
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas J. Booth
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia and
| | - Gnana D. Ardhanari
- Division of Pediatric Cardiac Critical Care Medicine, Children’s Heart Institute, Memorial Hermann Hospital, University of Texas Health McGovern Medical School, Houston, Texas, USA
| | - Jill M. Thompson
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia and
| | - L.K. Metthew Lam
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Department of Medicine and
| | - Jacob E. Till
- Division of Hematology-Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark V. Mai
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Garrett Keim
- Division of Pediatric Critical Care, Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia and
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J. McKeone
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Patrick Lahni
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Brian M. Varisco
- Section of Critical Care, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Wanding Zhou
- Center for Computational and Genomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Erica L. Carpenter
- Division of Hematology-Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Department of Medicine and
- Center for Translational Lung Biology and
- Center for Clinical Epidemiology and Biostatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nilam S. Mangalmurti
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Department of Medicine and
- Center for Translational Lung Biology and
| |
Collapse
|
45
|
Tang X, Guo J, Qi F, Rezaei MJ. Role of non-coding RNAs and exosomal non-coding RNAs in vasculitis: A narrative review. Int J Biol Macromol 2024; 261:129658. [PMID: 38266857 DOI: 10.1016/j.ijbiomac.2024.129658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
A category of very uncommon systemic inflammatory blood vessel illnesses known as vasculitides. The pathogenesis and etiology of vasculitis are still poorly known. Despite all of the progress made in understanding the genetics and causes behind vasculitis, there is still more to learn. Epigenetic dysregulation is a significant contributor to immune-mediated illnesses, and epigenetic aberrancies in vasculitis are becoming more widely acknowledged. Less than 2 % of the genome contains protein-encoding DNA. Studies have shown that a variety of RNAs originating from the non-coding genome exist. Long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) have attracted the most attention in recent years as they are becoming more and more important regulators of different biological processes, such as diseases of the veins. Extracellular vehicles (EVs) such as exosomes, are membrane-bound vesicular structures that break free either during programmed cell death, such as apoptosis, pyroptosis, and necroptosis or during cell activation. Exosomes may be involved in harmful ways in inflammation, procoagulation, autoimmune reactions, endothelial dysfunction/damage, intimal hyperplasia and angiogenesis, all of which may be significant in vasculitis. Herein, we summarized various non-coding RNAs that are involved in vasculitides pathogenesis. Moreover, we highlighted the role of exosomes in vasculitides.
Collapse
Affiliation(s)
- Xiuming Tang
- Department of Cardiology, The affiliated hospital to Changchun University of Chinise Medicine, Changchun, Jilin 130021, China.
| | - Jiajuan Guo
- Department of Cardiology, The affiliated hospital to Changchun University of Chinise Medicine, Changchun, Jilin 130021, China
| | - Feng Qi
- Department of Cardiology, The affiliated hospital to Changchun University of Chinise Medicine, Changchun, Jilin 130021, China
| | - Mohammad J Rezaei
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
46
|
Haybar H, Hadi H, Purrahman D, Mahmoudian-Sani MR, Saki N. Emerging roles of HOTAIR lncRNA in the pathogenesis and prognosis of cardiovascular diseases. Biomark Med 2024; 18:203-219. [PMID: 38411079 DOI: 10.2217/bmm-2023-0368] [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] [Indexed: 02/28/2024] Open
Abstract
Highlights HOTAIR, a long noncoding RNA, plays a role in the regulation of proteins involved in the pathogenesis of cardiovascular disease. Furthermore, it has been identified as a biomarker of this type of disease. Several factors and cells contribute to atherosclerosis, a progressive disease. However, the prognosis of HOTAIR in this disease varies depending on the path in which it plays a role. For this condition, there is no single prognosis to consider.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hakimeh Hadi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Daryush Purrahman
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
47
|
Fan A, Gao M, Tang X, Jiao M, Wang C, Wei Y, Gong Q, Zhong J. HMGB1/RAGE axis in tumor development: unraveling its significance. Front Oncol 2024; 14:1336191. [PMID: 38529373 PMCID: PMC10962444 DOI: 10.3389/fonc.2024.1336191] [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: 11/10/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
High mobility group protein 1 (HMGB1) plays a complex role in tumor biology. When released into the extracellular space, it binds to the receptor for advanced glycation end products (RAGE) located on the cell membrane, playing an important role in tumor development by regulating a number of biological processes and signal pathways. In this review, we outline the multifaceted functions of the HMGB1/RAGE axis, which encompasses tumor cell proliferation, apoptosis, autophagy, metastasis, and angiogenesis. This axis is instrumental in tumor progression, promoting tumor cell proliferation, autophagy, metastasis, and angiogenesis while inhibiting apoptosis, through pivotal signaling pathways, including MAPK, NF-κB, PI3K/AKT, ERK, and STAT3. Notably, small molecules, such as miRNA-218, ethyl pyruvate (EP), and glycyrrhizin exhibit the ability to inhibit the HMGB1/RAGE axis, restraining tumor development. Therefore, a deeper understanding of the mechanisms of the HMGB1/RAGE axis in tumors is of great importance, and the development of inhibitors targeting this axis warrants further exploration.
Collapse
Affiliation(s)
- Anqi Fan
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Mengxiang Gao
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
48
|
Lu T, Lahousse L, Wijnant S, Chen J, Brusselle GG, van Hoek M, Zillikens MC. The AGE-RAGE axis associates with chronic pulmonary diseases and smoking in the Rotterdam study. Respir Res 2024; 25:85. [PMID: 38336742 PMCID: PMC10858545 DOI: 10.1186/s12931-024-02698-1] [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: 09/04/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) and asthma associate with high morbidity and mortality. High levels of advanced glycation end products (AGEs) were found in tissue and plasma of COPD patients but their role in COPD and asthma is unclear. METHODS In the Rotterdam Study (n = 2577), AGEs (by skin autofluorescence (SAF)), FEV1 and lung diffusing capacity (DLCOc and DLCOc /alveolar volume [VA]) were measured. Associations of SAF with asthma, COPD, GOLD stage, and lung function were analyzed using logistic and linear regression adjusted for covariates, followed by interaction and stratification analyses. sRAGE and EN-RAGE associations with COPD prevalence were analyzed by logistic regression. RESULTS SAF associated with COPD prevalence (OR = 1.299 [1.060, 1.591]) but not when adjusted for smoking (OR = 1.106 [0.89, 1.363]). SAF associated with FEV1% predicted (β=-3.384 [-4.877, -1.892]), DLCOc (β=-0.212 [-0.327, -0.097]) and GOLD stage (OR = 4.073, p = 0.001, stage 3&4 versus 1). Stratified, the association between SAF and FEV1%predicted was stronger in COPD (β=-6.362 [-9.055, -3.670]) than non-COPD (β=-1.712 [-3.306, -0.118]). Association of SAF with DLCOc and DLCOc/VA were confined to COPD (β=-0.550 [-0.909, -0.191]; β=-0.065 [-0.117, -0.014] respectively). SAF interacted with former smoking and COPD prevalence for associations with lung function. Lower sRAGE and higher EN-RAGE associated with COPD prevalence (OR = 0.575[0.354, 0.931]; OR = 1.778[1.142, 2.768], respectively). CONCLUSIONS Associations between SAF, lung function and COPD prevalence were strongly influenced by smoking. SAF associated with COPD severity and its association with lung function was more prominent within COPD. These results fuel further research into interrelations and causality between SAF, smoking and COPD. TAKE-HOME MESSAGE Skin AGEs associated with prevalence and severity of COPD and lung function in the general population with a stronger effect in COPD, calling for further research into interrelations and causality between SAF, smoking and COPD.
Collapse
Affiliation(s)
- Tianqi Lu
- Department of Internal Medicine, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lies Lahousse
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Sara Wijnant
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Jinluan Chen
- Department of Internal Medicine, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015GD, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Guy G Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mandy van Hoek
- Department of Internal Medicine, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015GD, Rotterdam, The Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015GD, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Pasupulati AK, Nagati V, Paturi ASV, Reddy GB. Non-enzymatic glycation and diabetic kidney disease. VITAMINS AND HORMONES 2024; 125:251-285. [PMID: 38997166 DOI: 10.1016/bs.vh.2024.01.002] [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: 07/14/2024]
Abstract
Chronic diabetes leads to various complications including diabetic kidney disease (DKD). DKD is a major microvascular complication and the leading cause of morbidity and mortality in diabetic patients. Varying degrees of proteinuria and reduced glomerular filtration rate are the cardinal clinical manifestations of DKD that eventually progress into end-stage renal disease. Histopathologically, DKD is characterized by renal hypertrophy, mesangial expansion, podocyte injury, glomerulosclerosis, and tubulointerstitial fibrosis, ultimately leading to renal replacement therapy. Amongst the many mechanisms, hyperglycemia contributes to the pathogenesis of DKD via a mechanism known as non-enzymatic glycation (NEG). NEG is the irreversible conjugation of reducing sugars onto a free amino group of proteins by a series of events, resulting in the formation of initial Schiff's base and an Amadori product and to a variety of advanced glycation end products (AGEs). AGEs interact with cognate receptors and evoke aberrant signaling cascades that execute adverse events such as oxidative stress, inflammation, phenotypic switch, complement activation, and cell death in different kidney cells. Elevated levels of AGEs and their receptors were associated with clinical and morphological manifestations of DKD. In this chapter, we discussed the mechanism of AGEs accumulation, AGEs-induced cellular and molecular events in the kidney and their impact on the pathogenesis of DKD. We have also reflected upon the possible options to curtail the AGEs accumulation and approaches to prevent AGEs mediated adverse renal outcomes.
Collapse
Affiliation(s)
- Anil K Pasupulati
- Department of Biochemistry, University of Hyderabad, Hyderabad, India.
| | - Veerababu Nagati
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
| | - Atreya S V Paturi
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
| | - G Bhanuprakash Reddy
- Department of Biochemistry, ICMR-National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
50
|
Ye D, Miyoshi A, Ushitani T, Kadoya M, Igeta M, Konishi K, Shoji T, Yasuda K, Kitaoka S, Yagi H, Kuroda E, Yamamoto Y, Cheng J, Koyama H. RAGE in circulating immune cells is fundamental for hippocampal inflammation and cognitive decline in a mouse model of latent chronic inflammation. Brain Behav Immun 2024; 116:329-348. [PMID: 38142917 DOI: 10.1016/j.bbi.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Latent chronic inflammation has been proposed as a key mediator of multiple derangements in metabolic syndrome (MetS), which are increasingly becoming recognized as risk factors for age-related cognitive decline. However, the question remains whether latent chronic inflammation indeed induces brain inflammation and cognitive decline. METHODS A mouse model of latent chronic inflammation was constructed by a chronic subcutaneous infusion of low dose lipopolysaccharide (LPS) for four weeks. A receptor for advanced glycation end products (RAGE) knockout mouse, a chimeric myeloid cell specific RAGE-deficient mouse established by bone marrow transplantation and a human endogenous secretory RAGE (esRAGE) overexpressing adenovirus system were utilized to examine the role of RAGE in vivo. The cognitive function was examined by a Y-maze test, and the expression level of genes was determined by quantitative RT-PCR, western blot, immunohistochemical staining, or ELISA assays. RESULTS Latent chronic inflammation induced MetS features in C57BL/6J mice, which were associated with cognitive decline and brain inflammation characterized by microgliosis, monocyte infiltration and endothelial inflammation, without significant changes in circulating cytokines including TNF-α and IL-1β. These changes as well as cognitive impairment were rescued in RAGE knockout mice or chimeric mice lacking RAGE in bone marrow cells. P-selectin glycoprotein ligand-1 (PSGL-1), a critical adhesion molecule, was induced in circulating mononuclear cells in latent chronic inflammation in wild-type but not RAGE knockout mice. These inflammatory changes and cognitive decline induced in the wild-type mice were ameliorated by an adenoviral increase in circulating esRAGE. Meanwhile, chimeric RAGE knockout mice possessing RAGE in myeloid cells were still resistant to cognitive decline and brain inflammation. CONCLUSIONS These findings indicate that RAGE in inflammatory cells is necessary to mediate stimuli of latent chronic inflammation that cause brain inflammation and cognitive decline, potentially by orchestrating monocyte activation via regulation of PSGL-1 expression. Our results also suggest esRAGE-mediated inflammatory regulation as a potential therapeutic option for cognitive dysfunction in MetS with latent chronic inflammation.
Collapse
Affiliation(s)
- Dasen Ye
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Akio Miyoshi
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Tomoe Ushitani
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Manabu Kadoya
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Masataka Igeta
- Department of Biostatistics, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Kosuke Konishi
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Takuhito Shoji
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Koubun Yasuda
- Department of Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Shiho Kitaoka
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Hideshi Yagi
- Department of Anatomy and Cell Biology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Etsushi Kuroda
- Department of Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Jidong Cheng
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan; Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan.
| |
Collapse
|