1
|
Xuan L, Ren L, Kang X, Chang R, Zhang W, Gong L, Liu L. Clusterin ameliorates diabetic atherosclerosis by suppressing macrophage pyroptosis and activation. Front Pharmacol 2025; 16:1536132. [PMID: 40337510 PMCID: PMC12055819 DOI: 10.3389/fphar.2025.1536132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Background It has been demonstrated that clusterin (CLU) is a protective protein involved in a variety of diseases and disorders. However, the role of CLU in diabetic atherosclerosis is not elucidative. The objective of this study is to investigate the role of CLU in diabetic atherosclerosis and the molecular mechanisms. Method In in vivo experiments, Clu knockout and overexpressed murine models were used to investigate the role of Clu in diabetic atherosclerosis. Atherosclerotic plaque formation was determined by hematoxylin-eosin (H&E) staining and Oil Red O staining. F4/80 and CD68 levels were determined by immunohistochemical staining. Transmission electron microscopy was used to observe changes in cell pyroptosis morphology. NLRP3 and IL-1β levels were determined by Western blot and immunofluorescence staining. In in vitro experiments, TNF-α, IL-6, and IL-1β levels in THP-1 derived macrophages were determined by real-time qPCR and ELISA. Results We found that Clu-overexpression reduced while Clu knockout promoted atherosclerotic plaque formation, macrophage infiltration and inflammatory factor expression in mouse aortic plaques. Consistently, CLU overexpression inhibits the production of TNF-α, IL-6, and IL-1β in THP-1 derived macrophages. Moreover, Clu inhibited the release of inflammatory factors and macrophage pyroptosis in diabetic atherosclerosis murine models. Conclusion Our study revealed that CLU could ameliorate diabetic atherosclerosis via suppressing inflammatory factors release and pyroptosis of macrophage. CLU may be a promising therapeutic target for diabetic atherosclerosis.
Collapse
Affiliation(s)
- Lingling Xuan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lulu Ren
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoxu Kang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Rui Chang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Wen Zhang
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lili Gong
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Lihong Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
2
|
Du X, Chen Z, Shui W. Clusterin: structure, function and roles in disease. Int J Med Sci 2025; 22:887-896. [PMID: 39991767 PMCID: PMC11843140 DOI: 10.7150/ijms.107159] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/11/2025] [Indexed: 02/25/2025] Open
Abstract
Clusterin (CLU) is a glycoprotein that exists in various forms in cells, including nuclear, cytoplasmic, and secreted types. The relative molecular weight of CLU varies significantly due to differences in glycosylation and cleavage. Although CLU is commonly present in mammalian tissues and body fluids, its expression levels differ markedly under physiological and pathological conditions. The existence forms and molecular sizes of CLU in cells vary greatly, contributing to its diverse functions. For example, CLU can participate in the occurrence and development of neurological, fibrotic, and metabolic diseases by regulating cell endocytosis, apoptosis, and other processes. This article will review the structural characteristics, basic functions, and potential regulatory mechanisms of CLU protein in physiological and pathological processes.
Collapse
Affiliation(s)
- Xing Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Zhongyao Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Wei Shui
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
3
|
McGranaghan P, Pallinger É, Fekete N, Maurovich-Horvát P, Drobni Z, Merkely B, Menna L, Buzás EI, Hegyesi H. Modeling the Impact of Extracellular Vesicle Cargoes in the Diagnosis of Coronary Artery Disease. Biomedicines 2024; 12:2682. [PMID: 39767589 PMCID: PMC11727391 DOI: 10.3390/biomedicines12122682] [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: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Objectives: We aimed to assess the relationship among circulating extracellular vesicles (EVs), hypoxia-related proteins, and the conventional risk factors of life-threatening coronary artery disease (CAD) to find more precise novel biomarkers. Methods: Patients were categorized based on coronary CT angiography. Patients with a Segment Involvement Score > 5 were identified as CAD patients. Individuals with a Segment Involvement Score < 5 were considered control subjects. The characterization of EVs and analysis of the plasma concentration of growth differentiation factor-15 were performed using multicolor or bead-based flow cytometry. The plasma protein levels of glycogen phosphorylase, muscle form, clusterin, and carboxypeptidase N subunit 1 were determined using an enzyme-linked immunosorbent assay. Multiple logistic regression was used to determine the association of the biomarkers with the CAD outcome after accounting for established risk factors. The analysis was built in three steps: first, we included the basic clinical and laboratory variables (Model 1), then we integrated the plasma protein values (Model 2), and finally, we complemented it with the circulating EV pattern (Model 3). To assess the discrimination value of the models, an area under (AUC) the receiver operating curve was calculated and compared across the three models. Results: The area under the curve (AUC) values were 0.68, 0.77, and 0.84 in Models 1, 2, and 3, respectively. The variables with the greatest impact on the AUC values were hemoglobin (0.2 (0.16-0.26)) in Model 1, carboxypeptidase N subunit 1 (0.12 (0.09-0.14)) in Model 2, and circulating CD41+/CD61+ EVs (0.31 (0.15-0.5)) in Model 3. A correlation analysis showed a significant impact of circulating CD41+/CD61+ platelet-derived EVs (p = 0.03, r = -0.4176) in Model 3. Conclusions: Based on our results, the circulating EV profile can be used as a supportive biomarker, along with the conventional laboratory markers of CAD, and it enables a more sensitive, non-invasive diagnostic analysis of CAD.
Collapse
Affiliation(s)
- Peter McGranaghan
- Biomarker Department, Charité—Universitätsmedizin, 10117 Berlin, Germany
| | - Éva Pallinger
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary
| | - Nóra Fekete
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary
| | | | - Zsófia Drobni
- Medical Imaging Centre, Semmelweis University, 1085 Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 1085 Budapest, Hungary
| | - Luigi Menna
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary
| | - Edit I. Buzás
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN-SU Translational Extracellular Vesicle Research Group, 1085 Budapest, Hungary
- HCEMM-SU Extracellular Vesicle Research Group, 1085 Budapest, Hungary
| | - Hargita Hegyesi
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
4
|
Sarkar S, Prasanna VS, Das P, Suzuki H, Fujihara K, Kodama S, Sone H, Sreedhar R, Velayutham R, Watanabe K, Arumugam S. The onset and the development of cardiometabolic aging: an insight into the underlying mechanisms. Front Pharmacol 2024; 15:1447890. [PMID: 39391689 PMCID: PMC11464448 DOI: 10.3389/fphar.2024.1447890] [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: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024] Open
Abstract
Metabolic compromise is crucial in aggravating age-associated chronic inflammation, oxidative stress, mitochondrial damage, increased LDL and triglycerides, and elevated blood pressure. Excessive adiposity, hyperglycemia, and insulin resistance due to aging are associated with elevated levels of damaging free radicals, inducing a proinflammatory state and hampering immune cell activity, leading to a malfunctioning cardiometabolic condition. The age-associated oxidative load and redox imbalance are contributing factors for cardiometabolic morbidities via vascular remodelling and endothelial damage. Recent evidence has claimed the importance of gut microbiota in maintaining regular metabolic activity, which declines with chronological aging and cardiometabolic comorbidities. Genetic mutations, polymorphic changes, and environmental factors strongly correlate with increased vulnerability to aberrant cardiometabolic changes by affecting key physiological pathways. Numerous studies have reported a robust link between biological aging and cardiometabolic dysfunction. This review outlines the scientific evidence exploring potential mechanisms behind the onset and development of cardiovascular and metabolic issues, particularly exacerbated with aging.
Collapse
Affiliation(s)
- Sulogna Sarkar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Vani S. Prasanna
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Pamelika Das
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Hiroshi Suzuki
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoru Kodama
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Remya Sreedhar
- School of Pharmacy, Sister Nivedita University, Kolkata, West Bengal, India
| | - Ravichandiran Velayutham
- Director, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Kenichi Watanabe
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| |
Collapse
|
5
|
Jalilvand A, Ireland M, Collins C, Kellett W, Strassel S, Tamer R, Wahl W, Wisler J. Obesity is associated with improved early survival but increased late mortality in surgical patients with Sepsis: A propensity matched analysis. J Trauma Acute Care Surg 2024; 97:233-241. [PMID: 38480496 PMCID: PMC11531704 DOI: 10.1097/ta.0000000000004316] [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: 07/26/2024]
Abstract
BACKGROUND While obesity is a risk factor for postoperative complications, its impact following sepsis is unclear. The primary objective of this study was to evaluate the association between obesity and mortality following admission to the surgical intensive care unit (SICU) with sepsis. METHODS We conducted a single center retrospective review of SICU patients grouped into obese (n = 766, body mass index ≥30 kg/m 2 ) and nonobese (n = 574; body mass index, 18-29.9 kg/m 2 ) cohorts. Applying 1:1 propensity matching for age, sex, comorbidities, sequential organ failure assessment, and transfer status, demographic data, comorbidities, and sepsis presentation were compared between groups. Primary outcomes included in-hospital and 90-day mortality, ICU length of stay, need for mechanical ventilation (IMV) and renal replacement therapy (RRT). p < 0.05 was considered significant. RESULTS Obesity associates with higher median ICU length of stay (8.2 vs. 5.6, p < 0.001), need for IMV (76% vs. 67%, p = 0.001), ventilator days (5 vs. 4, p < 0.004), and RRT (23% vs. 12%, p < 0.001). In-hospital (29% vs. 18%, p < 0.0001) and 90-day mortality (34% vs. 24%, p = 0.0006) was higher for obese compared with nonobese groups. Obesity independently predicted need for IMV (odds ratio [OR], 1.6; 95% confidence interval [CI], 1.2-2.1), RRT (OR, 2.2; 95% CI, 1.5-3.1), in-hospital (OR, 2.1; 95% CI, 1.5-2.8), and 90-day mortality (HR, 1.4; 95% CI, 1.1-1.8), after adjusting for sequential organ failure assessment, age, sex, and comorbidities. Comparative survival analyses demonstrate a paradoxical early survival benefit for obese patients followed by a rapid decline after 7 days (logrank p = 0.0009). CONCLUSION Obesity is an independent risk factor for 90-day mortality for surgical patients with sepsis, but its impact appeared later in hospitalization. Understanding differences in systemic responses between these cohorts may be important for optimizing critical care management. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
Affiliation(s)
- Anahita Jalilvand
- From the Division of Trauma, Critical Care, and Burn (A.J., C.C., W.K., R.T., W.W., J.W.), Ohio State University, Columbus, Ohio; University Kentucky College of Medicine (M.I.), Lexington, Kentucky; and Atrium Health (S.S.), Charlotte, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Chen C, Xie L, Zhang M, Shama, Cheng KKY, Jia W. The interplay between the muscle and liver in the regulation of glucolipid metabolism. J Mol Cell Biol 2024; 15:mjad073. [PMID: 38095440 PMCID: PMC11078061 DOI: 10.1093/jmcb/mjad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/24/2023] [Indexed: 05/09/2024] Open
Affiliation(s)
- Cheng Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Liping Xie
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Mingliang Zhang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Shama
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
7
|
Lu J, Wang Z, Zhang J, Jiao F, Zou C, Han L, Jiang G. Causal association of blood lipids with all-cause and cause-specific mortality risk: a Mendelian randomization study. J Lipid Res 2024; 65:100528. [PMID: 38458338 PMCID: PMC10993189 DOI: 10.1016/j.jlr.2024.100528] [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: 07/28/2023] [Revised: 02/03/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
Dyslipidemia has long been implicated in elevating mortality risk; yet, the precise associations between lipid traits and mortality remained undisclosed. Our study aimed to explore the causal effects of lipid traits on both all-cause and cause-specific mortality. One-sample Mendelian randomization (MR) with linear and nonlinear assumptions was conducted in a cohort of 407,951 European participants from the UK Biobank. Six lipid traits, consisting of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), and lipoprotein(a), were included to investigate the causal associations with mortality. Two-sample MR was performed to replicate the association between each lipid trait and all-cause mortality. Univariable MR results showed that genetically predicted higher ApoA1 was significantly associated with a decreased all-cause mortality risk (HR[95% CI]:0.93 [0.89-0.97], P value = 0.001), which was validated by the two-sample MR analysis. Higher lipoprotein(a) was associated with an increased risk of all-cause mortality (1.03 [1.01-1.04], P value = 0.002). Multivariable MR confirmed the direct causal effects of ApoA1 and lipoprotein(a) on all-cause mortality. Meanwhile, nonlinear MR found no evidence for nonlinearity between lipids and all-cause mortality. Our examination into cause-specific mortality revealed a suggestive inverse association between ApoA1 and cancer mortality, a significant positive association between lipoprotein(a) and cardiovascular disease mortality, and a suggestive positive association between lipoprotein(a) and digestive disease mortality. High LDL-C was associated with an increased risk of cardiovascular disease mortality but a decreased risk of neurodegenerative disease mortality. The findings suggest that implementing interventions to raise ApoA1 and decrease lipoprotein(a) levels may improve overall health outcomes and mitigate cancer and digestive disease mortality.
Collapse
Affiliation(s)
- Jiawen Lu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiaying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Jiao
- Guangzhou Centre for Applied Mathematics, Guangzhou University, Guangzhou, Guangdong, China
| | - Chenfeng Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liyuan Han
- Department of Global Health, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Chen Z, Zeng H, Huang Q, Lin C, Li X, Sun S, Liu JP. Increased GPC4 and clusterin associated with insulin resistance in patients with PCOS. Endocr Connect 2024; 13:e230428. [PMID: 38251963 PMCID: PMC10959037 DOI: 10.1530/ec-23-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/22/2024] [Indexed: 01/23/2024]
Abstract
The aim of the study was to investigate the changes in serum glypican 4 (GPC4) and clusterin (CLU) levels in patients with polycystic ovary syndrome (PCOS) as well as their correlation with sex hormones and metabolic parameters. A total of 40 PCOS patients and 40 age-matched healthy women were selected. Serum GPC4 and CLU levels were compared between the PCOS and control groups, and binary logistic regression was used to analyze the relative risk of PCOS at different tertiles of serum GPC4 and CLU concentrations. Stepwise linear regression was used to identify the factors influencing serum GPC4 and CLU levels in PCOS patients. Serum GPC4 (1.82 ± 0.49 vs 1.30 ± 0.61 ng/mL, P < 0.001) and CLU (468.79 ± 92.85 vs 228.59 ± 82.42 µg/mL, P < 0.001) were significantly higher in PCOS patients than in healthy women after adjustment for body mass index (BMI). In the PCOS group, serum GPC4 was positively correlated with follicle-stimulating hormone, fasting plasma glucose (FPG), fasting insulin (FINS), homeostatic model assessment of insulin resistance (HOMA-IR), triglyceride, and CLU (P < 0.05), whereas serum CLU was positively correlated with BMI, FPG, FINS, and HOMA-IR (P < 0.05). Multiple stepwise linear regression analysis showed that HOMA-IR was independently associated with serum GPC4, and BMI and HOMA-IR were independently associated with CLU (P < 0.05). Serum GPC4 and CLU levels were significantly higher in PCOS patients than in healthy women, suggesting that GPC4 and CLU may be markers associated with insulin resistance in women with PCOS.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haixia Zeng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiulan Huang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cuiping Lin
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shaohua Sun
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian-ping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Kostopoulou E, Kalavrizioti D, Davoulou P, Papachristou E, Sinopidis X, Fouzas S, Dassios T, Gkentzi D, Kyriakou SI, Karatza A, Dimitriou G, Goumenos D, Spiliotis BE, Plotas P, Papasotiriou M. Monocyte Chemoattractant Protein-1 (MCP-1), Activin-A and Clusterin in Children and Adolescents with Obesity or Type-1 Diabetes Mellitus. Diagnostics (Basel) 2024; 14:450. [PMID: 38396489 PMCID: PMC10887959 DOI: 10.3390/diagnostics14040450] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/04/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
UNLABELLED Inflammation plays a crucial role in diabetes and obesity through macrophage activation. Macrophage chemoattractant protein-1 (MCP-1), activin-A, and clusterin are chemokines with known roles in diabetes and obesity. The aim of this study is to investigate their possible diagnostic and/or early prognostic values in children and adolescents with obesity and type-1 diabetes mellitus (T1DM). METHODS We obtained serum samples from children and adolescents with a history of T1DM or obesity, in order to measure and compare MCP-1, activin-A, and clusterin concentrations. RESULTS Forty-three subjects were included in each of the three groups (controls, T1DM, and obesity). MCP-1 values were positively correlated to BMI z-score. Activin-A was increased in children with obesity compared to the control group. A trend for higher values was detected in children with T1DM. MCP-1 and activin-A levels were positively correlated. Clusterin levels showed a trend towards lower values in children with T1DM or obesity compared to the control group and were negatively correlated to renal function. CONCLUSIONS The inflammation markers MCP-1, activin-A, and clusterin are not altered in children with T1DM. Conversely, obesity in children is positively correlated to serum MCP-1 values and characterized by higher activin-A levels, which may reflect an already established systematic inflammation with obesity since childhood.
Collapse
Affiliation(s)
- Eirini Kostopoulou
- Division of Pediatric Endocrinology, Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (E.K.); (B.E.S.)
| | - Dimitra Kalavrizioti
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Panagiota Davoulou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Evangelos Papachristou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Xenophon Sinopidis
- Department of Pediatric Surgery, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Sotirios Fouzas
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Theodore Dassios
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Despoina Gkentzi
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Stavroula Ioanna Kyriakou
- Department of Pediatric Surgery, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Ageliki Karatza
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Gabriel Dimitriou
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Dimitrios Goumenos
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Bessie E. Spiliotis
- Division of Pediatric Endocrinology, Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (E.K.); (B.E.S.)
| | - Panagiotis Plotas
- Department of Speech and Language Therapy, School of Health Rehabilitation Sciences, University of Patras, 26504 Patras, Greece;
| | - Marios Papasotiriou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| |
Collapse
|
10
|
Abstract
Obesity is epidemic and of great concern because of its comorbid and costly inflammatory-driven complications. Extensive investigations in mice have elucidated highly coordinated, well-balanced interactions between adipocytes and immune cells in adipose tissue that maintain normal systemic metabolism in the lean state, while in obesity, proinflammatory changes occur in nearly all adipose tissue immune cells. Many of these changes are instigated by adipocytes. However, less is known about obesity-induced adipose-tissue immune cell alterations in humans. Upon high-fat diet feeding, the adipocyte changes its well-known function as a metabolic cell to assume the role of an immune cell, orchestrating proinflammatory changes that escalate inflammation and progress during obesity. This transformation is particularly prominent in humans. In this review, we (a) highlight a leading and early role for adipocytes in promulgating inflammation, (b) discuss immune cell changes and the time course of these changes (comparing humans and mice when possible), and (c) note how reversing proinflammatory changes in most types of immune cells, including adipocytes, rescues adipose tissue from inflammation and obese mice from insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Pennsylvania State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA;
| | - Tuo Deng
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| |
Collapse
|
11
|
Duan S, Qin N, Pi J, Sun P, Gao Y, Liu L, Li Z, Li Y, Shi L, Gao Q, Qiu Y, Tang S, Wang CH, Chen TY, Wang ST, Young KC, Sun HY. Antagonizing apolipoprotein J chaperone promotes proteasomal degradation of mTOR and relieves hepatic lipid deposition. Hepatology 2023; 78:1182-1199. [PMID: 37013405 DOI: 10.1097/hep.0000000000000185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/26/2022] [Indexed: 04/05/2023]
Abstract
BACKGROUND AND AIMS Overnutrition-induced activation of mammalian target of rapamycin (mTOR) dysregulates intracellular lipid metabolism and contributes to hepatic lipid deposition. Apolipoprotein J (ApoJ) is a molecular chaperone and participates in pathogen-induced and nutrient-induced lipid accumulation. This study investigates the mechanism of ApoJ-regulated ubiquitin-proteasomal degradation of mTOR, and a proof-of-concept ApoJ antagonist peptide is proposed to relieve hepatic steatosis. APPROACH AND RESULTS By using omics approaches, upregulation of ApoJ was found in high-fat medium-fed hepatocytes and livers of patients with NAFLD. Hepatic ApoJ level associated with the levels of mTOR and protein markers of autophagy and correlated positively with lipid contents in the liver of mice. Functionally, nonsecreted intracellular ApoJ bound to mTOR kinase domain and prevented mTOR ubiquitination by interfering FBW7 ubiquitin ligase interaction through its R324 residue. In vitro and in vivo gain-of-function or loss-of-function analysis further demonstrated that targeting ApoJ promotes proteasomal degradation of mTOR, restores lipophagy and lysosomal activity, thus prevents hepatic lipid deposition. Moreover, an antagonist peptide with a dissociation constant (Kd) of 2.54 µM interacted with stress-induced ApoJ and improved hepatic pathology, serum lipid and glucose homeostasis, and insulin sensitivity in mice with NAFLD or type II diabetes mellitus. CONCLUSIONS ApoJ antagonist peptide might be a potential therapeutic against lipid-associated metabolic disorders through restoring mTOR and FBW7 interaction and facilitating ubiquitin-proteasomal degradation of mTOR.
Collapse
Affiliation(s)
- Shuangdi Duan
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Nong Qin
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Jiayi Pi
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Pei Sun
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Yating Gao
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Lamei Liu
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Zenghui Li
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Ya Li
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Liyang Shi
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
| | - Qiang Gao
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Ye Qiu
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Songqing Tang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology of College of Biology, Hunan University, Changsha, China
| | - Chun-Hsiang Wang
- Division of Gastroenterology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Tzu-Ying Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sin-Tian Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kung-Chia Young
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Yu Sun
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
12
|
Charisis S, Lin H, Ray R, Joehanes R, Beiser AS, Levy D, Seshadri S, Sargurupremraj M, Satizabal CL. Obesity impacts the expression of Alzheimer's disease-related genes: The Framingham Heart Study. Alzheimers Dement 2023; 19:3496-3505. [PMID: 36811231 PMCID: PMC10435662 DOI: 10.1002/alz.12954] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 02/24/2023]
Abstract
INTRODUCTION We investigated associations of obesity with the expression of Alzheimer's disease (AD)-related genes in a large community-based cohort. METHODS The sample consisted of 5619 participants from the Framingham Heart Study. Obesity metrics included body mass index (BMI) and waist-to-hip ratio (WHR). Gene expression was measured for a set of 74 AD-related genes, derived by integrating genome-wide association study results with functional genomics data. RESULTS Obesity metrics were associated with the expression of 21 AD-related genes. The strongest associations were observed with CLU, CD2AP, KLC3, and FCER1G. Unique associations were noted with TSPAN14, SLC24A4 for BMI, and ZSCAN21, BCKDK for WHR. After adjustment for cardiovascular risk factors, 13 associations remained significant for BMI and 8 for WHR. Dichotomous obesity metrics exhibited unique associations with EPHX2 for BMI, and with TSPAN14 for WHR. DISCUSSION Obesity was associated with AD-related gene expression; these findings shed light on the molecular pathways linking obesity to AD.
Collapse
Affiliation(s)
- Sokratis Charisis
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Honghuang Lin
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Roshni Ray
- Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Roby Joehanes
- The Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
| | - Alexa S Beiser
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Muralidharan Sargurupremraj
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| |
Collapse
|
13
|
Clusterin Plasma Concentrations Are Decreased in Sepsis and Inversely Correlated with Established Markers of Inflammation. Diagnostics (Basel) 2022; 12:diagnostics12123010. [PMID: 36553017 PMCID: PMC9776480 DOI: 10.3390/diagnostics12123010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022] Open
Abstract
Clusterin is a multifunctional protein that is recognized to mediate cellular stress response associated with organ failure, systemic inflammation, and metabolic alterations. The aim of this study was to determine the value of clusterin as a clinical biomarker in critical ill patients with or without sepsis. We analyzed clusterin plasma concentrations in 200 critically ill patients (133 with sepsis, 67 without sepsis) on admission to the medical intensive care unit (ICU). The results were compared with 66 healthy controls. Clusterin plasma concentration was significantly elevated in critically ill patients compared to healthy subjects. Clusterin levels were significantly higher in non-septic ICU patients than in patients with sepsis. Clusterin correlated inversely with routinely used biomarkers of inflammatory response. Furthermore, clusterin levels were higher in ICU patients with pre-existing obesity and type 2 diabetes. Clusterin was not associated with disease severity, organ failure, or mortality in the ICU. This study highlights significantly elevated clusterin levels in critically ill patients, predominantly in non-sepsis conditions, and associates circulating clusterin to inflammatory and metabolic dysfunctions.
Collapse
|
14
|
Bradley D, Smith AJ, Blaszczak A, Shantaram D, Bergin SM, Jalilvand A, Wright V, Wyne KL, Dewal RS, Baer LA, Wright KR, Stanford KI, Needleman B, Brethauer S, Noria S, Renton D, Joseph JJ, Lovett-Racke A, Liu J, Hsueh WA. Interferon gamma mediates the reduction of adipose tissue regulatory T cells in human obesity. Nat Commun 2022; 13:5606. [PMID: 36153324 PMCID: PMC9509397 DOI: 10.1038/s41467-022-33067-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
Decreased adipose tissue regulatory T cells contribute to insulin resistance in obese mice, however, little is known about the mechanisms regulating adipose tissue regulatory T cells numbers in humans. Here we obtain adipose tissue from obese and lean volunteers. Regulatory T cell abundance is lower in obese vs. lean visceral and subcutaneous adipose tissue and associates with reduced insulin sensitivity and altered adipocyte metabolic gene expression. Regulatory T cells numbers decline following high-fat diet induction in lean volunteers. We see alteration in major histocompatibility complex II pathway in adipocytes from obese patients and after high fat ingestion, which increases T helper 1 cell numbers and decreases regulatory T cell differentiation. We also observe increased expression of inhibitory co-receptors including programmed cell death protein 1 and OX40 in visceral adipose tissue regulatory T cells from patients with obesity. In human obesity, these global effects of interferon gamma to reduce regulatory T cells and diminish their function appear to instigate adipose inflammation and suppress adipocyte metabolism, leading to insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
- Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Alan J Smith
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Alecia Blaszczak
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Stephen M Bergin
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anahita Jalilvand
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Valerie Wright
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kathleen L Wyne
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Revati S Dewal
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Lisa A Baer
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Katherine R Wright
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristin I Stanford
- Diabetes and Metabolism Research Center, Division of Physiology and Cell Biology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Stacy Brethauer
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - David Renton
- Center for Minimally Invasive Surgery, Department of General Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Joshua J Joseph
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Amy Lovett-Racke
- Department of Microbial Immunity and Infection, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Joey Liu
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
15
|
The Antidiabetic Activities of Neocryptotanshinone: Screened by Molecular Docking and Related to the Modulation of PTP1B. Nutrients 2022; 14:nu14153031. [PMID: 35893885 PMCID: PMC9330310 DOI: 10.3390/nu14153031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
The aim of this study was to provide a practical experimental basis for the development of Neocryptotanshinone (NCTS) as an effective hypoglycemic drug and a theoretical method for the rapid screening of natural compounds with hypoglycemic effects. Molecular docking was used to screen the most suitable ligand. Hematoxylin and eosin, immunohistochemical staining, enzyme-linked immunosorbent assay and Western Blotting approved the hypoglycemic effect of NCTS. According to the free energy of binding, among 180 active compounds from the Traditional Chinese Medicine Integrated Database, NCTS was finally chose for investigation its hypoglycemic effects. In db/db mice, NCTS significantly reduced body weight and plasma glucose, improved glucose tolerance and levels of fasting plasma glucose and glycated hemoglobin A1c, and decreased insulin resistance after six-week administration. NCTS restored the pathological state in the liver of db/db mice and significantly decreased protein tyrosine phosphatase 1B (PTP1B) expression in the liver and muscle of db/db mice, which is related to the regulatory effect of NCTS on insulin receptor substrate 1. In conclusion, we successfully explored the hypoglycemic effect of NCTS in db/db mice via regulating the expression of PTP1B.
Collapse
|
16
|
Li P, Cong Y, Zhang W, Wang L, Ren L, Li X, Yang S, Zhang Z, Li G, Liu L. Simultaneous quantification of apolipoproteins A-I, E, and J in human plasma by LC-MS/MS for clinical application to diabetes mellitus complicated with cardiovascular disease. RSC Adv 2022; 12:16763-16771. [PMID: 35754896 PMCID: PMC9170378 DOI: 10.1039/d2ra02840a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/29/2022] [Indexed: 11/25/2022] Open
Abstract
Apolipoproteins (Apos) play an important role in regulating plasma lipid concentration. Complex disorders of Apos are highly related with diabetes mellitus, cardiovascular and other diseases. Direct measures of lipoprotein fractions for risk assessment suffer from inaccuracy in the dyslipidemia and pathological states. Therefore, a reliable precise assay will be of high clinical utility. LC-MS/MS methods with multiple reaction monitoring modes have proven suitable for multiplexed quantification. We aimed to develop a simple, cost-effective and amenable LC-MS/MS assay for quantification of ApoA-I, ApoE and ApoJ in human plasma. Standards were constructed from substitute matrix and proteotypic peptides for external calibration and corresponding stable isotope labeled peptides were added as internal standards to remove matrix effects. Analytical validation of the assay included the assessment of linearity, accuracy (RE: −3.02% to 5.32%), intra-assay precision (RSD: 2.50% to 6.56%), inter-assay precision (RSD: 0.78% to 6.68%), spiking recovery rate (accuracy: 87.17% to 112.71%), matrix effect (accuracy: 88.03% to 114.87%), and reproducibility and repeatability of sample preparation (RSD: 1.95% to 7.26%). The performance of proteotypic peptides ApoA-I, ApoE and ApoJ was sufficient for triplex quantitation within a linear range from 16.26 to 1626.41 pmol mL−1, 1.03 to 103.35 pmol mL−1 and 0.86 to 86.46 pmol mL−1 respectively. For all quantified peptides, the determination coefficient (R2) was >0.997. Besides, the validated LC-MS/MS method has been successfully applied to the quantification of plasma samples in diabetes mellitus and cardiovascular diseases. We anticipate that this assay may provide an alternative method for future clinical applications. Simultaneous quantification of apolipoproteins A-I, E, and J in human plasma by LC-MS/MS.![]()
Collapse
Affiliation(s)
- Pengfei Li
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Yuting Cong
- Shanghai AB Sciex Analytical Instrument Trading Co. Ltd 518 North Fuquan Road, Changning District Shanghai China
| | - Wen Zhang
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Lefeng Wang
- Heart Center of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Lulu Ren
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Xin Li
- Heart Center of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Song Yang
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Zhiyong Zhang
- Heart Center of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| | - Guoqing Li
- Shanghai AB Sciex Analytical Instrument Trading Co. Ltd 518 North Fuquan Road, Changning District Shanghai China
| | - Lihong Liu
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University No. 8 Gongti South Road, Chaoyang District Beijing China
| |
Collapse
|
17
|
Zhang Q, Meng XH, Qiu C, Shen H, Zhao Q, Zhao LJ, Tian Q, Sun CQ, Deng HW. Integrative analysis of multi-omics data to detect the underlying molecular mechanisms for obesity in vivo in humans. Hum Genomics 2022; 16:15. [PMID: 35568907 PMCID: PMC9107154 DOI: 10.1186/s40246-022-00388-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Obesity is a complex, multifactorial condition in which genetic play an important role. Most of the systematic studies currently focuses on individual omics aspect and provide insightful yet limited knowledge about the comprehensive and complex crosstalk between various omics levels. SUBJECTS AND METHODS Therefore, we performed a most comprehensive trans-omics study with various omics data from 104 subjects, to identify interactions/networks and particularly causal regulatory relationships within and especially those between omic molecules with the purpose to discover molecular genetic mechanisms underlying obesity etiology in vivo in humans. RESULTS By applying differentially analysis, we identified 8 differentially expressed hub genes (DEHGs), 14 differentially methylated regions (DMRs) and 12 differentially accumulated metabolites (DAMs) for obesity individually. By integrating those multi-omics biomarkers using Mendelian Randomization (MR) and network MR analyses, we identified 18 causal pathways with mediation effect. For the 20 biomarkers involved in those 18 pairs, 17 biomarkers were implicated in the pathophysiology of obesity or related diseases. CONCLUSIONS The integration of trans-omics and MR analyses may provide us a holistic understanding of the underlying functional mechanisms, molecular regulatory information flow and the interactive molecular systems among different omic molecules for obesity risk and other complex diseases/traits.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Community Nursing, School of Nursing and Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Xiang-He Meng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qi Zhao
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Lan-Juan Zhao
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chang-Qing Sun
- Department of Community Nursing, School of Nursing and Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
- Department of Social Medicine and Health Management, College of Public Health, Zhengzhou University, High-Tech Development Zone of States, Zhengzhou, 450001, Henan, People's Republic of China
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
18
|
Sanda GM, Toma L, Barbalata T, Moraru OE, Niculescu LS, Sima AV, Stancu CS. Clusterin, paraoxonase 1, and myeloperoxidase alterations induce high-density lipoproteins dysfunction and contribute to peripheral artery disease; aggravation by type 2 diabetes mellitus. Biofactors 2022; 48:454-468. [PMID: 34741558 DOI: 10.1002/biof.1800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022]
Abstract
Peripheral artery disease (PAD) is an atherosclerotic disorder affecting arteries of the lower limbs, the major risk factors including dyslipidemia and diabetes mellitus (DM). We aimed to identify alterations of the proteins in high-density lipoproteins (HDL) associated with HDL dysfunction in PAD patients. HDL2 and HDL3 were isolated from plasma of PAD patients with/without DM (PAD-DM/PAD) and healthy subjects (N). Apolipoprotein AI (ApoAI), ApoAII, ApoCIII, clusterin (CLU), paraoxonase 1 (PON1), myeloperoxidase (MPO), and ceruloplasmin (CP) were measured in HDL2 /HDL3 and plasma. Oxidation and glycation of the analyzed proteins were assessed as malondialdehyde-protein adducts (MDA) and advanced glycation end-products (AGE), respectively. The anti-inflammatory effect of HDL3 was estimated as its potential to reduce monocyte adhesion to tumor necrosis factor α-activated endothelial cells. We show that in PAD patients compared to N subjects: (i) HDL2 presented increased levels of MDA-PON1, AGE-PON1, AGE-ApoAI, ApoAII, ApoCIII, and CP levels, and decreased PON1 levels; (ii) HDL3 had increased levels of MDA- and AGE-CLU and -ApoAI, MDA-PON1, ApoCIII, CLU, MPO, CP, and reduced PON1 levels. All these alterations were exacerbated by DM. These changes were more pronounced in HDL3 , which had reduced anti-inflammatory potential in PAD and became pro-inflammatory in PAD-DM. In PAD patients' plasma, CLU levels and MPO specific activity increased, while PON1 specific activity decreased. In conclusion, HDL function is altered in PAD patients due to multiple modifications of associated proteins that are aggravated by DM. Plasma CLU, MPO, and PON1 could constitute indicators of HDL dysfunction and contribute to risk stratification in PAD patients.
Collapse
Affiliation(s)
- Gabriela M Sanda
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Laura Toma
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Teodora Barbalata
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Oriana E Moraru
- Emergency Clinical Hospital "Prof. Dr. Agrippa Ionescu", Ilfov County, Romania
| | - Loredan S Niculescu
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Anca V Sima
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Camelia S Stancu
- Lipidomics Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
19
|
Andújar-Vera F, García-Fontana C, Sanabria-de la Torre R, González-Salvatierra S, Martínez-Heredia L, Iglesias-Baena I, Muñoz-Torres M, García-Fontana B. Identification of Potential Targets Linked to the Cardiovascular/Alzheimer's Axis through Bioinformatics Approaches. Biomedicines 2022; 10:389. [PMID: 35203598 PMCID: PMC8962298 DOI: 10.3390/biomedicines10020389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 12/23/2022] Open
Abstract
The identification of common targets in Alzheimer's disease (AD) and cardiovascular disease (CVD) in recent years makes the study of the CVD/AD axis a research topic of great interest. Besides aging, other links between CVD and AD have been described, suggesting the existence of common molecular mechanisms. Our study aimed to identify common targets in the CVD/AD axis. For this purpose, genomic data from calcified and healthy femoral artery samples were used to identify differentially expressed genes (DEGs), which were used to generate a protein-protein interaction network, where a module related to AD was identified. This module was enriched with the functionally closest proteins and analyzed using different centrality algorithms to determine the main targets in the CVD/AD axis. Validation was performed by proteomic and data mining analyses. The proteins identified with an important role in both pathologies were apolipoprotein E and haptoglobin as DEGs, with a fold change about +2 and -2, in calcified femoral artery vs healthy artery, respectively, and clusterin and alpha-2-macroglobulin as close interactors that matched in our proteomic analysis. However, further studies are needed to elucidate the specific role of these proteins, and to evaluate its function as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Francisco Andújar-Vera
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Raquel Sanabria-de la Torre
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Sheila González-Salvatierra
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Iván Iglesias-Baena
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), 18012 Granada, Spain;
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18012 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (B.G.-F.)
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio of Granada, 18016 Granada, Spain
- CIBERFES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
20
|
Bradley D, Xu A, Hsueh WA. Editorial: The Immunomodulatory Roles of Adipocytes. Front Immunol 2022; 12:827281. [PMID: 35003144 PMCID: PMC8732371 DOI: 10.3389/fimmu.2021.827281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
21
|
Mirzaeicheshmeh E, Zerrweck C, Centeno-Cruz F, Baca-Peynado P, Martinez-Hernandez A, García-Ortiz H, Contreras-Cubas C, Salas-Martínez MG, Saldaña-Alvarez Y, Mendoza-Caamal EC, Barajas-Olmos F, Orozco L. Alterations of DNA methylation during adipogenesis differentiation of mesenchymal stem cells isolated from adipose tissue of patients with obesity is associated with type 2 diabetes. Adipocyte 2021; 10:493-504. [PMID: 34699309 PMCID: PMC8555535 DOI: 10.1080/21623945.2021.1978157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022] Open
Abstract
Adipogenesis regulation is crucial for mature adipocyte function. In obesity, a major driver of type 2 diabetes (T2D), this process is disrupted and remains poorly characterized. Here we identified altered DNA methylation profiles in diabetic obese patients, during three adipocytes differentiation stages. We isolated mesenchymal cells from visceral adipose tissue of obese patients with and without T2D to analyse DNA methylation profiles at 0, 3, and 18 days of ex vivo differentiation and documented their impact on gene expression. Methylation and gene expression were analysed with EPIC and Clarion S arrays, respectively. Patients with T2D had epigenetic alterations in all the analysed stages, and these were mainly observed in genes important in adipogenesis, insulin resistance, cell death programming, and immune effector processes. Importantly, at 3 days, we found six-fold more methylated CpG alterations than in the other stages. This is the first study to document epigenetic markers that persist through all three adipogenesis stages and their impact on gene expression, which could be a cellular metabolic memory involved in T2D. Our data provided evidence that, throughout the adipogenesis process, alterations occur in methylation that might impact mature adipocyte function, cause tissue malfunction, and potentially, lead to the development of T2D.
Collapse
Affiliation(s)
- Elaheh Mirzaeicheshmeh
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Carlos Zerrweck
- Clínica de Obesidad del Hospital General Tláhuac, SSA, Mexico City, Mexico
- Facultad De Medicina, Alta Especialidad En Cirugía Bariatrica, Unam, Mexico City, Mexico
| | - Federico Centeno-Cruz
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Paulina Baca-Peynado
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Angélica Martinez-Hernandez
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Humberto García-Ortiz
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Cecilia Contreras-Cubas
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | | | - Yolanda Saldaña-Alvarez
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | | | - Francisco Barajas-Olmos
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| | - Lorena Orozco
- Immunogenomics and Metabolic Disease Laboratory, Instituto Nacional De Medicina Genómica, Ss, Mexico City, Mexico
| |
Collapse
|
22
|
Zeng S, Pan Y, Liu F, Yin J, Jiang M, Long Y, Zhao X, Lash GE, Yang H. Role of clusterin in the regulation of trophoblast development and preeclampsia. Biochem Biophys Res Commun 2021; 583:128-134. [PMID: 34735874 DOI: 10.1016/j.bbrc.2021.10.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/21/2022]
Abstract
Preeclampsia (PE) threatens the safety of mothers and fetuses, and its pathogenesis is still unclear. Our previous study has found the relationship between PE and serum Clusterin (CLU). This study aimed to investigate the role of CLU on PE. Firstly, levels of CLU in serum and placental tissue from PE patients and healthy pregnancies were compared. Then, RNA sequencing, cell counting kit-8, matrigel invasion, cell apoptosis, and angiogenesis assay were performed to evaluate the role of CLU on primary isolation trophoblast cells. We found the expression of CLU was increased before the clinical syndrome occurred, whereas its level was positively related to the severity of PE. CLU significantly inhibited the expression of matrix metalloproteinase-9 and Vimentin and enhanced E-cadherin to inhibit epithelial-mesenchymal transition of trophoblast cells, further reducing its migration and invasion. Our results suggested that CLU may play a role in regulating trophoblast invasion and migration during placental development, which may be one of the risk factors for PE.
Collapse
Affiliation(s)
- Shanshui Zeng
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yue Pan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Fei Liu
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jiaye Yin
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Min Jiang
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yan Long
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xueqin Zhao
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China
| | - Gendie E Lash
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Hongling Yang
- Department of Laboratory, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
23
|
Chen X, Ma H, Gao Y, Jin Y, Ning W, Hou Y, Su J. Long non-coding RNA AC012668 suppresses non-alcoholic fatty liver disease by competing for microRNA miR-380-5p with lipoprotein-related protein LRP2. Bioengineered 2021; 12:6738-6747. [PMID: 34511037 PMCID: PMC8806601 DOI: 10.1080/21655979.2021.1960463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by high morbidity. Although long noncoding RNAs (lncRNAs) are known to have a role in NAFLD pathogenesis, the identified lncRNA types are limited. In this study, NAFLD models were established in vitro and in vivo using free fatty acid-treated LO2 cells and high-fat diet-fed mice, respectively. Microarray data were downloaded from the Gene Expression Omnibus database, and AC012668 was selected for further analysis. Cell viability and apoptosis were measured using Cell Counting Kit 8 and flow cytometry assays. RNA expression was detected using reverse transcription-quantitative polymerase chain reaction. Triglyceride (TG) content and lipid deposition were detected using enzyme-linked immunosorbent assay and Oil-Red O staining. Western blotting was used to visualize protein expression. Starbase and TargetScan were used to predict the target miRNA and gene, and the predictions were verified through RNA pull-down and luciferase reporter assays. AC012668 expression levels were significantly suppressed in NAFLD models, whereas AC012668 overexpression inhibited lipogenesis-related gene (SCD1, SREBP1, FAS) expression and TG/lipid accumulation in vitro. Subsequently, miR-380-5p was predicted and verified to target AC012668, and its expression was notably increased in the NAFLD cell model. Moreover, transfection of miR-380-5p antagonized the effects of AC012668 on lipid formation and accumulation. LRP2 was confirmed to be the target gene of miR-380-5p and was downregulated in the NAFLD cell model. Silencing LRP2 reversed the effects of the miR-380-5p inhibitor on lipid formation and accumulation. AC012668 inhibited NAFLD progression via the miR-380-5p/LRP2 axis. These findings may provide a novel strategy against NAFLD.
Collapse
Affiliation(s)
- Xiaomeng Chen
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| | - Hong Ma
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yan Gao
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| | - Ye Jin
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| | - Wei Ning
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| | - Yue Hou
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| | - Jianrong Su
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, China
| |
Collapse
|
24
|
Sun HY, Chen TY, Tan YC, Wang CH, Young KC. Sterol O-acyltransferase 2 chaperoned by apolipoprotein J facilitates hepatic lipid accumulation following viral and nutrient stresses. Commun Biol 2021; 4:564. [PMID: 33980978 PMCID: PMC8115332 DOI: 10.1038/s42003-021-02093-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 04/06/2021] [Indexed: 11/08/2022] Open
Abstract
The risks of non-alcoholic fatty liver disease (NAFLD) include obese and non-obese stresses such as chronic hepatitis C virus (HCV) infection, but the regulatory determinants remain obscure. Apolipoprotein J (ApoJ) served as an ER-Golgi contact-site chaperone near lipid droplet (LD), facilitating HCV virion production. We hypothesized an interplay between hepatic ApoJ, cholesterol esterification and lipid deposit in response to NAFLD inducers. Exposures of HCV or free-fatty acids exhibited excess LDs along with increased ApoJ expression, whereas ApoJ silencing alleviated hepatic lipid accumulation. Both stresses could concomitantly disperse Golgi, induce closer ApoJ and sterol O-acyltransferase 2 (SOAT2) contacts via the N-terminal intrinsically disordered regions, and increase cholesteryl-ester. Furthermore, serum ApoJ correlated positively with cholesterol and low-density lipoprotein levels in normal glycaemic HCV patients, NAFLD patients and in mice with steatosis. Taken together, hepatic ApoJ might activate SOAT2 to supply cholesteryl-ester for lipid loads, thus providing a therapeutic target of stress-induced steatosis.
Collapse
Affiliation(s)
- Hung-Yu Sun
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, China
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, Hunan University, Changsha, China
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Ying Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ching Tan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Hsiang Wang
- Division of Gastroenterology, Tainan Municipal Hospital, Tainan, Taiwan
| | - Kung-Chia Young
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
25
|
Abelleira E, Peñaloza MA, Jerkovich F, Bueno F, Pitoia F. Dynamic risk allows us to adequately select patients with differentiated thyroid cancer who do not require radioiodine treatment. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:2359-3997000000355. [PMID: 33939904 PMCID: PMC10065312 DOI: 10.20945/2359-3997000000355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The treatment of patients with differentiated thyroid cancer (DTC) was modified in the last decade towards a more individualized approach according to the risk of recurrence (RR). We compared the outcomes of patients with low and intermediate RR (LRR and IRR) who received or did not receive radioiodine remnant ablation (RRA) after assessing the dynamic risk. METHODS We included 307 DTC patients with LRR and IRR submitted to total thyroidectomy. All patients were reclassified according to the dynamic risk stratification (low or high). Patients with high dynamic risk received RRA (141 patients). RESULTS LRR patients who received RRA presented a frequency of structural incomplete response (SIR) of 5% at the end of the follow-up, compared to 2% in those who did not receive it (p=0.353). IRR patients treated with RRA had a frequency of SIR of 22%, compared to 5% in patients without RRA (p=0.008). CONCLUSION This study demonstrates the usefulness of dynamic risk assessment to decide RRA in a cohort with a long-term follow-up. The lower prevalence of SIR at the end of the follow-up in patients who did not receive RRA highlights the adequate selection of those who would not benefit from RRA, even with an intermediate risk of recurrence.
Collapse
Affiliation(s)
- Erika Abelleira
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | | | - Fernando Jerkovich
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Fernanda Bueno
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Fabián Pitoia
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina,
| |
Collapse
|
26
|
Höpfinger A, Berghoff M, Karrasch T, Schmid A, Schäffler A. Systematic Quantification of Neurotrophic Adipokines RBP4, PEDF, and Clusterin in Human Cerebrospinal Fluid and Serum. J Clin Endocrinol Metab 2021; 106:e2239-e2250. [PMID: 33484131 DOI: 10.1210/clinem/dgaa983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Indexed: 02/08/2023]
Abstract
CONTEXT Data on the presence/quantification of the neurotrophic adipokines retinol-binding protein-4 (RBP4), clusterin, and pigment epithelium-derived factor (PEDF) in human cerebrospinal fluid (CSF) are scarce and migration of these adipokines across of the blood-brain barrier (BBB) is uncertain. OBJECTIVE This work aimed to quantify RBP4, PEDF, and clusterin in paired serum and CSF samples of patients undergoing neurological evaluation. METHODS A total of 268 patients (109 male, 159 female) were included. Adipokine serum and CSF concentrations were measured by enzyme-linked immunosorbent assay in duplicate. RESULTS RBP4 was abundant in serum (mean, 31.9 ± 24.2 μg/mL). The serum concentrations were approximately 145 times higher than in CSF (CSF to serum RBP4 ratio, 8.2 ± 4.3 × 10-3). PEDF was detectable in serum (mean, 30.2 ± 11.7 μg/mL) and concentrations were approximately 25 times higher than in CSF (CSF to serum PEDF ratio, 42.3 ± 15.6 × 10-3). Clusterin serum concentrations were abundant with mean levels of 346.0 ± 114.6 μg/mL, which were approximately 40 times higher than CSF levels (CSF to serum clusterin ratio, 29.6 ± 23.4 × 10-3). RBP4 and PEDF serum levels correlated positively with CSF levels, which were increased in overweight/obese patients and in type 2 diabetic patients. The CSF concentrations of all 3 adipokines increased with BBB dysfunction. RBP4 in CSF correlated positively with inflammatory parameters. In detail, only RBP4 showed the kinetics and associations that are mandatory for a putative mediator of the fat-brain axis. CONCLUSION RBP4, PEDF, and clusterin are permeable to the BBB and increase with the measure of BBB dysfunction. RBP4 represents an inflammatory neurotrophic adipokine and is a promising mediator of the fat-brain axis.
Collapse
Affiliation(s)
- Alexandra Höpfinger
- Department of Internal Medicine III, Giessen University Hospital, Gießen, Germany
| | - Martin Berghoff
- Department of Neurology, Giessen University Hospital, Gießen, Germany
| | - Thomas Karrasch
- Department of Internal Medicine III, Giessen University Hospital, Gießen, Germany
| | - Andreas Schmid
- Department of Internal Medicine III, Giessen University Hospital, Gießen, Germany
| | - Andreas Schäffler
- Department of Internal Medicine III, Giessen University Hospital, Gießen, Germany
| |
Collapse
|
27
|
Wittwer J, Bradley D. Clusterin and Its Role in Insulin Resistance and the Cardiometabolic Syndrome. Front Immunol 2021; 12:612496. [PMID: 33717095 PMCID: PMC7946829 DOI: 10.3389/fimmu.2021.612496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The cardiometabolic syndrome involves a clustering of metabolic and cardiovascular factors which increase the risk of patients developing both Type 2 Diabetes Mellitus and cardio/cerebrovascular disease. Although the mechanistic underpinnings of this link remain uncertain, key factors include insulin resistance, excess visceral adiposity, atherogenic dyslipidemia, and endothelial dysfunction. Of these, a state of resistance to insulin action in overweight/obese patients appears to be central to the pathophysiologic process. Given the increasing prevalence of obesity-related Type 2 Diabetes, coupled with the fact that cardiovascular disease is the number one cause of mortality in this patient population, a more thorough understanding of the cardiometabolic syndrome and potential options to mitigate its risk is imperative. Inherent in the pathogenesis of insulin resistance is an underlying state of chronic inflammation, at least partly in response to excess adiposity. Within obese adipose tissue, an immunomodulatory shift occurs, involving a preponderance of pro-inflammatory immune cells and cytokines/adipokines, along with antigen presentation by adipocytes. Therefore, various adipokines differentially expressed by obese adipocytes may have a significant effect on cardiometabolism. Clusterin is a molecular chaperone that is widely produced by many tissues throughout the body, but is also preferentially overexpressed by obese compared lean adipocytes and relates strongly to multiple components of the cardiometabolic syndrome. Herein, we summarize the known and potential roles of circulating and adipocyte-specific clusterin in cardiometabolism and discuss potential further investigations to determine if clusterin is a viable target to attenuate both metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer Wittwer
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
28
|
Werida RH, El-Gharbawy NM, Mostafa TM. Circulating IL-6, clusterin and irisin in obese subjects with different grades of obesity: association with insulin resistance and sexual dimorphism. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:126-136. [PMID: 33905632 PMCID: PMC10065324 DOI: 10.20945/2359-3997000000336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective There are discrepancies about the relationship of IL-6, clusterin and irisin with obesity and obesity associated insulin resistance and also about their sexual dimorphism. This study aimed at evaluating the circulating levels of IL-6, clusterin and irisin in obese subjects of both sexes who had different grades of obesity and examining their sexual dimorphism and their association with insulin resistance. Methods This study included 176 non-diabetic subjects of both sexes who were classified according to their sex into two groups; the male and the female groups. The male group (88 men) was classified according to BMI into; group 1 (22 lean men), group 2 (22 class I obese men), group 3 (22 class II obese men) and group 4 (22 class III obese men). The female group (88 women) was classified according to BMI exactly as the male group. Metabolic parameters, IL-6, clusterin, and irisin levels were measured. Data were analyzed by ANOVA test, post hoc Tukey's test and independent t-test. Pearson correlation was used to assess the association between variables. Results In obese subjects of both sexes, circulating IL-6, clusterin and irisin levels were significantly elevated and positively correlated with HOMA-IR. Obese males showed significantly higher HOMA-IR, IL-6, clusterin and irisin levels than obese females. Conclusion Obesity in both sexes, especially in males was associated with high levels of IL-6, clusterin and irisin and worsened the metabolic pattern. Circulating IL-6, clusterin and irisin may represent possible therapeutic targets for insulin resistance in obese subjects.
Collapse
Affiliation(s)
- Rehab H Werida
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Damanhur University, Damanhur, Egypt
| | - Nashwa M El-Gharbawy
- Department of Internal Medicine, Diabetes & Endocrinology Unit, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Tarek M Mostafa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt,
| |
Collapse
|
29
|
Rodríguez-Rivera C, Garcia MM, Molina-Álvarez M, González-Martín C, Goicoechea C. Clusterin: Always protecting. Synthesis, function and potential issues. Biomed Pharmacother 2021; 134:111174. [DOI: 10.1016/j.biopha.2020.111174] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
|
30
|
Sato N. The Emerging Role of the Apolipoproteins APOE and APOJ in the Interaction Between Diabetes and Alzheimer Disease. J Clin Endocrinol Metab 2020; 105:5902587. [PMID: 32895708 DOI: 10.1210/clinem/dgaa570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/17/2020] [Indexed: 01/17/2023]
Affiliation(s)
- Naoyuki Sato
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
31
|
Bradley D. Clusterin as a Potential Biomarker of Obesity-Related Alzheimer's Disease Risk. Biomark Insights 2020; 15:1177271920964108. [PMID: 33110346 PMCID: PMC7555556 DOI: 10.1177/1177271920964108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/03/2023] Open
Abstract
Over 35% of the adult US population is obese. In turn, excess adiposity increases the risk of multiple complications including type 2 diabetes (T2D), insulin resistance, and cardiovascular disease; yet, obesity also independently heightens risk of Alzheimer's Disease (AD), even after adjusting for other important confounding risk factors including blood pressure, sociodemographics, cholesterol levels, smoking status, and Apolipoprotein E (ApoE) genotype. Among patients over the age of 65 with dementia, 37% have coexisting diabetes, and an estimated 7.3% of cases of AD are directly attributable to midlife obesity. Clusterin, also known as apolipoprotein J (ApoJ), is a multifunctional glycoprotein that acts as a molecular chaperone, assisting folding of secreted proteins. Clusterin has been implicated in several physiological and pathological states, including AD, metabolic disease, and cardiovascular disease. Despite long-standing interest in elucidating clusterin's relationship with amyloid beta (Aβ) aggregation/clearance and toxicity, significant knowledge gaps still exist. Altered clusterin expression and protein levels have been linked with cognitive and memory function, disrupted central nervous system lipid flux, as well as pathogenic brain structure; and its role in cardiometabolic disease suggests that it may be a link between insulin resistance, dyslipidemia, and AD. Here, we briefly highlight clusterin's relevance to AD by presenting existing evidence linking clusterin to AD and cardiometabolic disease, and discussing its potential utility as a biomarker for AD in the presence of obesity-related metabolic disease.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
32
|
Ha J, Moon MK, Kim H, Park M, Cho SY, Lee J, Lee JY, Kim E. Plasma Clusterin as a Potential Link Between Diabetes and Alzheimer Disease. J Clin Endocrinol Metab 2020; 105:5860166. [PMID: 32561922 DOI: 10.1210/clinem/dgaa378] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/13/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Plasma clusterin, a promising biomarker of Alzheimer disease (AD), has been associated with diabetes mellitus (DM). However, clusterin has not been investigated considering a relationship with both DM and AD. In this study, we aimed to investigate the individual and interactive relationships of plasma clusterin levels with both diseases. DESIGN Cross-sectional observation study. METHODS We classified participants by the severity of cognitive (normal cognition, mild cognitive impairment [MCI], and AD) and metabolic (healthy control, prediabetes, and DM) impairments. We evaluated the cognitive and metabolic functions of the participants with neuropsychological assessments, brain magnetic resonance imaging, and various blood tests, to explore potential relationships with clusterin. RESULTS Plasma clusterin levels were higher in participants with AD and metabolic impairment (prediabetes and DM). A two-way ANCOVA revealed no synergistic, but an additive effect of AD and DM on clusterin. Clusterin was negatively correlated with cognitive scores. It was also associated with metabolic status indicated by glycated hemoglobin A1c (HbA1c), the Homeostatic Model Assessment for Insulin Resistance index, and fasting C-peptide. It showed correlations between medial temporal atrophy and periventricular white matter lesions, indicating neurodegeneration and microvascular insufficiency, respectively. Further mediation analysis to understand the triadic relationship between clusterin, AD, and DM revealed that the association between DM and AD was significant when clusterin is considered as a mediator of their relationship. CONCLUSIONS Clusterin is a promising biomarker of DM as well as of AD. Additionally, our data suggest that clusterin may have a role in linking DM with AD as a potential mediator.
Collapse
Affiliation(s)
- Junghee Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Kyong Moon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsun Park
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jimin Lee
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry and Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eosu Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Seo JA, Kang MC, Yang WM, Hwang WM, Kim SS, Hong SH, Heo JI, Vijyakumar A, Pereira de Moura L, Uner A, Huang H, Lee SH, Lima IS, Park KS, Kim MS, Dagon Y, Willnow TE, Aroda V, Ciaraldi TP, Henry RR, Kim YB. Apolipoprotein J is a hepatokine regulating muscle glucose metabolism and insulin sensitivity. Nat Commun 2020; 11:2024. [PMID: 32332780 PMCID: PMC7181874 DOI: 10.1038/s41467-020-15963-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/02/2020] [Indexed: 12/24/2022] Open
Abstract
Crosstalk between liver and skeletal muscle is vital for glucose homeostasis. Hepatokines, liver-derived proteins that play an important role in regulating muscle metabolism, are important to this communication. Here we identify apolipoprotein J (ApoJ) as a novel hepatokine targeting muscle glucose metabolism and insulin sensitivity through a low-density lipoprotein receptor-related protein-2 (LRP2)-dependent mechanism, coupled with the insulin receptor (IR) signaling cascade. In muscle, LRP2 is necessary for insulin-dependent IR internalization, an initial trigger for insulin signaling, that is crucial in regulating downstream signaling and glucose uptake. Of physiologic significance, deletion of hepatic ApoJ or muscle LRP2 causes insulin resistance and glucose intolerance. In patients with polycystic ovary syndrome and insulin resistance, pioglitazone-induced improvement of insulin action is associated with an increase in muscle ApoJ and LRP2 expression. Thus, the ApoJ-LRP2 axis is a novel endocrine circuit that is central to the maintenance of normal glucose homeostasis and insulin sensitivity. Hepatokines are proteins secreted by the liver that can regulate whole body metabolism. Here the authors identify apolipoprotein J as a hepatokine that regulates muscle glucose metabolism and insulin resistance through a low-density lipoprotein receptor-related protein−2 mediated mechanism in mice.
Collapse
Affiliation(s)
- Ji A Seo
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Division of Endocrinology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Min-Cheol Kang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Research Group of Food Processing, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Korea
| | - Won-Mo Yang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Won Min Hwang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Division of Nephrology, Department of Internal Medicine, College of Medicine, Konyang University, Daejeon, Korea
| | - Sang Soo Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Soo Hyun Hong
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Columbia University, New York, NY, USA
| | - Jee-In Heo
- Division of Endocrinology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Achana Vijyakumar
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Leandro Pereira de Moura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,School of Applied Science, University of Campinas, Limeira, Brazil
| | - Aykut Uner
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hu Huang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,East Carolina University, East Carolina Diabetes and Obesity Institute, Greenville, NC, USA
| | - Seung Hwan Lee
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Inês S Lima
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.,Universidade Nova de Lisboa, Lisboa, Portugal
| | - Kyong Soo Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Min Seon Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Yossi Dagon
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Thomas E Willnow
- Molecular Cardiovascular Research, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Vanita Aroda
- Veterans Affairs San Diego Healthcare System (9111 G), San Diego, CA, 92161, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.,Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Theodore P Ciaraldi
- Veterans Affairs San Diego Healthcare System (9111 G), San Diego, CA, 92161, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Robert R Henry
- Veterans Affairs San Diego Healthcare System (9111 G), San Diego, CA, 92161, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Veyel D, Wenger K, Broermann A, Bretschneider T, Luippold AH, Krawczyk B, Rist W, Simon E. Biomarker discovery for chronic liver diseases by multi-omics - a preclinical case study. Sci Rep 2020; 10:1314. [PMID: 31992752 PMCID: PMC6987209 DOI: 10.1038/s41598-020-58030-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a major cause of liver fibrosis with increasing prevalence worldwide. Currently there are no approved drugs available. The development of new therapies is difficult as diagnosis and staging requires biopsies. Consequently, predictive plasma biomarkers would be useful for drug development. Here we present a multi-omics approach to characterize the molecular pathophysiology and to identify new plasma biomarkers in a choline-deficient L-amino acid-defined diet rat NASH model. We analyzed liver samples by RNA-Seq and proteomics, revealing disease relevant signatures and a high correlation between mRNA and protein changes. Comparison to human data showed an overlap of inflammatory, metabolic, and developmental pathways. Using proteomics analysis of plasma we identified mainly secreted proteins that correlate with liver RNA and protein levels. We developed a multi-dimensional attribute ranking approach integrating multi-omics data with liver histology and prior knowledge uncovering known human markers, but also novel candidates. Using regression analysis, we show that the top-ranked markers were highly predictive for fibrosis in our model and hence can serve as preclinical plasma biomarkers. Our approach presented here illustrates the power of multi-omics analyses combined with plasma proteomics and is readily applicable to human biomarker discovery.
Collapse
Affiliation(s)
- Daniel Veyel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Kathrin Wenger
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Andre Broermann
- Boehringer Ingelheim Pharma GmbH & Co. KG, CardioMetabolic Diseases Research, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Tom Bretschneider
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Andreas H Luippold
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Bartlomiej Krawczyk
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany
| | - Wolfgang Rist
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany.
| | - Eric Simon
- Boehringer Ingelheim Pharma GmbH & Co. KG, Computational Biology, Birkendorfer Str. 65, D-88397, Biberach Riss, Germany.
| |
Collapse
|
35
|
Ren L, Han F, Xuan L, Lv Y, Gong L, Yan Y, Wan Z, Guo L, Liu H, Xu B, Sun Y, Yang S, Liu L. Clusterin ameliorates endothelial dysfunction in diabetes by suppressing mitochondrial fragmentation. Free Radic Biol Med 2019; 145:357-373. [PMID: 31614179 DOI: 10.1016/j.freeradbiomed.2019.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022]
Abstract
Clusterin (CLU) is a stress-responding protein associated with cytoprotection in a broad range of pathological processes. However, clusterin's function in diabetes-induced endothelial dysfunction has not been defined. Herein, using two diabetes models, we investigated the role of clusterin in endothelial dysfunction triggered by diabetes and the molecular mechanisms involved. The results revealed that clusterin overexpression inhibited ICAM-1/VCAM-1 expression in aortas and improved endothelium-dependent vasodilatation in db/db diabetic mice and streptozotocin (STZ)-induced diabetes models. Consistently, in vitro, adenoviral clusterin overexpression reduced the expression of a range of pro-inflammatory cytokines and suppressed monocyte adhesion to endothelial cells subjected to high glucose and high palmitate. Further study indicated that clusterin overexpression mitigated mitochondrial excessive fission and reduced mitochondrial ROS production. Conversely, silencing clusterin aggravated mitochondrial fission and endothelial inflammatory activation in high glucose-exposed endothelial cells. Accumulating evidence indicates that impaired mitochondrial dynamics plays a considerable role in promoting endothelial dysfunction in diabetic subjects. Therefore, treatments targeting mitochondrial undue fission may be promising measures to prevent vascular complications of diabetes. Furthermore, AMP-activated protein kinase (AMPK) activation contributed to the modulation of mitochondrial dynamics executed by clusterin. Mechanistically, clusterin promoted the phosphorylation of AMPKα and its downstream target acetyl-CoA carboxylase (ACC), while the inhibition of AMPKα negated the improvement in mitochondrial dynamics provided by clusterin overexpression. Over all, these findings suggest that clusterin exerts beneficial effects in endothelial cells under diabetic conditions via inhibiting mitochondrial fragmentation mediated by AMPK.
Collapse
Affiliation(s)
- Lulu Ren
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Feifei Han
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lingling Xuan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yali Lv
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lili Gong
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yan Yan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zirui Wan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lifang Guo
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - He Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Benshan Xu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuan Sun
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Song Yang
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lihong Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|